GOLM1 is a Golgi membrane protein with a coding gene of 3,042 bp in full length. It is synthesized in the endoplasmic reticulum and primarily transported to the cis-Golgi apparatus. GOLM1 also functions as a secreted protein that can be detected in blood and urine. The protein is mainly expressed in epithelial cells and is upregulated in response to viral infection. It can be expressed in most human tissues, though predominantly in epithelial cells . When studying GOLM1 localization, immunofluorescence microscopy with Golgi markers is the recommended approach to confirm its subcellular distribution pattern.
The complete biological function of GOLM1 in cells and tissues remains partially characterized. Current evidence suggests GOLM1 plays an important role in supporting normal cell function. Methodologically, researchers should approach GOLM1 functional studies through both loss-of-function (siRNA, CRISPR knockout) and gain-of-function (overexpression) experiments to elucidate its physiological roles. The protein's secretory nature suggests it may have both intracellular and extracellular functions that should be investigated separately .
GOLM1 expression is notably upregulated in response to viral infection and shows significantly higher expression in tumor tissues compared to normal tissues across most cancer types (observed in 85.19% or 23/27 tumor types studied). This differential expression makes it a potentially valuable biomarker. When designing experiments to study GOLM1 regulation, researchers should include appropriate physiological stimuli such as viral components, inflammatory mediators, and analyze both transcriptional and post-transcriptional regulatory mechanisms .
GOLM1 appears to promote cancer progression through multiple mechanisms. Studies have linked increased GOLM1 expression with malignant biological characteristics. At the molecular level, GOLM1 can inhibit IL-12 production by dendritic cells, thus potentially promoting tumor progression by preventing T cell responses. It can also repress host innate immune processes, which may enhance viral replication (as observed with hepatitis C virus) and drive pathogenesis. Additionally, GOLM1 regulates the STAT3 pathway, which is upstream of PD-L1, suggesting a mechanism for immune checkpoint modulation. For experimental validation of these mechanisms, researchers should employ pathway inhibitors, co-immunoprecipitation assays, and immune cell co-culture systems .
GOLM1 expression correlates with specific cancer subtypes and genetic features. In uveal melanoma, higher GOLM1 expression was observed in epithelioid tumors compared to mixed tumors (p = 0.042), which aligns with the worse prognosis of epithelioid UVM. Additionally, UVM patients with monosomy chromosome 3 (a known poor prognostic indicator) exhibited higher GOLM1 levels than patients with disomy chromosome 3 (p = 0.017). When investigating such correlations, researchers should perform comprehensive genetic profiling alongside GOLM1 expression analysis to identify meaningful associations .
GOLM1 significantly impacts the tumor immune microenvironment. Gene set enrichment analysis of GOLM1-associated differentially expressed genes revealed enrichment in immune-related pathways. High GOLM1 expression correlates with increased infiltration of specific immune cell populations, particularly T cells. In UVM, the GOLM1-high (HEXP) group displayed significantly higher infiltration of type 1 T helper cells, CD8 T cells, natural killer T cells, T follicular helper cells, and CD4 T cells compared to the GOLM1-low (LEXP) group. When studying this aspect, researchers should employ single-cell RNA sequencing or spatial transcriptomics alongside flow cytometry to comprehensively map immune cell distributions and states in relation to GOLM1 expression .
GOLM1 expression exhibits strong positive correlations with immune checkpoint molecules. Immunohistochemistry analysis demonstrated that patients with high GOLM1 expression also displayed elevated levels of PD-1, PD-L1, CTLA-4, and IFN-γ. Specifically, statistical analysis revealed significant correlations between GOLM1 H-score and PD-L1 (R = 0.6065, p = 0.0028), CTLA-4 (R = 0.6834, p = 0.0003), and IFN-γ (R = 0.4521, p = 0.0303). These findings suggest GOLM1 may serve as a biomarker for immune checkpoint expression. When designing experiments to investigate these relationships, researchers should employ multiplexed immunofluorescence techniques and co-expression analysis at both protein and mRNA levels .
Analysis suggests GOLM1 expression can predict immunotherapy response. Subclass mapping analysis comparing GOLM1 expression groups with immunotherapy response signatures derived from melanoma cohorts treated with anti-PD-1 or anti-CTLA-4 revealed that UVM patients with high GOLM1 expression were predicted to be more sensitive to both PD-1/PD-L1 and CTLA4 therapies than those with lower GOLM1 expression. This finding was supported by the positive correlation between GOLM1 and checkpoint molecules. For clinical validation, researchers should analyze GOLM1 expression in pre-treatment biopsies from immunotherapy trials and correlate with treatment outcomes .
Multiple techniques can effectively measure GOLM1 expression in clinical samples. For mRNA analysis, quantitative RT-PCR and RNA sequencing are recommended. For protein detection, immunohistochemistry (IHC) with appropriate scoring systems (such as H-score) has proven effective. The search results describe using the median expression value to stratify patients into GOLM1-high and GOLM1-low groups for prognostic analysis. When implementing these methods, researchers should include appropriate controls and standardized protocols to ensure reproducibility. For circulating GOLM1, ELISA or other protein quantification methods can be applied to blood or urine samples .
For differential gene expression analysis related to GOLM1, researchers should first stratify samples based on GOLM1 expression levels. The "limma" package is recommended for identifying differentially expressed genes (DEGs) between GOLM1-high and GOLM1-low groups, with filtering criteria of fold-change >1 and p-value <0.01. Pathway enrichment analysis should be performed using tools such as "ClusterProfiler" with established gene set databases including KEGG, Gene Ontology (GO), and HALLMARK signatures from MSigDB. This approach revealed 531 DEGs between GOLM1 expression groups in UVM, with significant enrichment in immune-related pathways .
To analyze GOLM1's impact on immune cell infiltration, the single sample gene set enrichment analysis (ssGSEA) approach with the "GSVA" R package is recommended. Researchers should utilize established immunocyte signature gene sets (the search results mention using 28 immunocyte signatures) to calculate normalized enrichment scores (NES) for each sample. Statistical comparison of these scores between GOLM1-high and GOLM1-low groups can reveal differential immune cell infiltration patterns. This approach successfully identified significant differences in T cell subpopulations between GOLM1 expression groups across multiple independent cohorts .
GOLM1 interacts with several signaling pathways implicated in cancer progression. Evidence indicates GOLM1 regulates the STAT3 pathway, which is upstream of PD-L1, suggesting a mechanism for modulating immune checkpoint expression. Additionally, enrichment analysis of GOLM1-associated genes revealed connections to T cell receptor signaling, cytokine-cytokine receptor interaction, and antigen processing and presentation pathways. To investigate these interactions, researchers should employ phospho-proteomics, pathway inhibition experiments, and co-immunoprecipitation studies to establish direct versus indirect regulatory relationships .
The mechanisms driving GOLM1's differential expression between cancer and normal tissues require further investigation. While viral infection has been established as one trigger for GOLM1 upregulation, the search results don't fully elucidate the cancer-specific regulatory mechanisms. Researchers should investigate transcriptional regulation (promoter analysis, transcription factor binding), epigenetic modifications (DNA methylation, histone modifications), and post-transcriptional regulation (miRNA targeting) of GOLM1 in matched cancer and normal tissues. Chromosome 9q21.33, where the GOLM1 gene is located, should be analyzed for copy number alterations or structural variations in cancer .
Given GOLM1's correlation with immune checkpoint molecules and immunotherapy response prediction, GOLM1-targeted therapies might significantly impact the tumor immune microenvironment. Potential approaches include developing antibodies against GOLM1, small molecule inhibitors targeting GOLM1 function, or siRNA-based therapeutics. To investigate this experimentally, researchers should establish GOLM1 knockdown or knockout models and evaluate changes in immune cell infiltration, checkpoint molecule expression, and response to immune checkpoint blockade. Co-culture systems with immune cells would provide valuable insights into how GOLM1 modulation affects immune cell function and activation status .
GOLM1 expression analysis shows potential for integration into clinical decision-making through multiple applications. First, as a prognostic biomarker, it can help stratify patients' risk, with high expression consistently associated with poorer outcomes across cancer types (meta-analysis HR: 2.11, 95% CI: 1.30–3.43). Second, it could guide immunotherapy decisions, as patients with high GOLM1 expression are predicted to respond better to checkpoint inhibitors. To implement this clinically, standardized IHC or molecular testing protocols need development, with clearly defined cut-off values for stratification. Prospective clinical studies combining GOLM1 testing with treatment decision algorithms would be necessary for validation .
Validating GOLM1 as a therapeutic target requires a systematic approach. In vitro studies should establish the effects of GOLM1 inhibition on cancer cell proliferation, invasion, and migration across multiple cell lines. In vivo studies using xenograft or genetically engineered mouse models with GOLM1 modulation are essential to confirm its role in tumor growth and metastasis. Target specificity must be thoroughly assessed to minimize off-target effects, particularly given GOLM1's expression in normal tissues. Combination strategies with immunotherapies should be evaluated given the established correlations with immune checkpoints. Finally, biomarker development to identify patients most likely to benefit from GOLM1-targeted therapies would be crucial for clinical translation .
Technical variations in GOLM1 detection methods can significantly impact research reproducibility and clinical utility. For RNA-based methods, differences in sample collection, extraction methods, and platform selection (microarray vs. RNA-seq) can affect expression measurements. For protein detection, antibody selection, IHC protocols, and scoring systems introduce variability. The search results demonstrate using different cohorts (TCGA-UVM, GSE21138, GSE84976) and methodologies, yet achieving consistent findings regarding GOLM1's prognostic value, which suggests robust biological effects despite technical variations. Researchers should implement rigorous standardization, include appropriate controls, and validate findings across multiple technical platforms and independent cohorts to ensure reproducibility. For clinical implementation, standardized assays with established cut-offs would need development and validation .
The GOLM1 gene is located on chromosome 9 (9q21.33) in humans . It encodes a protein that is approximately 400 amino acids in length and has a molecular weight of around 73 kDa . The protein consists of a single transmembrane domain, with the N-terminus located in the cytoplasm and the C-terminus in the lumen of the Golgi apparatus .
GOLM1 is involved in the processing and transport of proteins synthesized in the rough endoplasmic reticulum . It is particularly important in the post-translational modification of proteins, including glycosylation, which is essential for the proper functioning of many proteins . The expression of GOLM1 is upregulated in response to viral infections, suggesting a role in the cellular response to viral pathogens .
GOLM1 has been implicated in various diseases, particularly those affecting the liver. It is overexpressed in liver diseases such as autoimmune hepatitis and liver cancer . Elevated levels of GOLM1 have been observed in the blood of patients with liver cancer, making it a potential biomarker for the early detection of this disease . Additionally, GOLM1 is overexpressed in prostate cancer and lung adenocarcinoma .
Recombinant GOLM1 is produced using recombinant DNA technology, where the GOLM1 gene is cloned into an expression vector and introduced into a host cell, typically E. coli or mammalian cells . The host cells then produce the GOLM1 protein, which can be purified and used for various research and diagnostic purposes . Recombinant GOLM1 is valuable for studying the protein’s function, interactions, and role in disease, as well as for developing diagnostic assays and potential therapeutic interventions .