GNG11 (Guanine Nucleotide-Binding Protein G(I)/G(S)/G(O) Subunit Gamma-11) is a gene encoding a 73-amino-acid protein critical for transmembrane signaling as part of the heterotrimeric G protein complex . It belongs to the G protein gamma subunit family, which interacts with beta subunits to regulate GTPase activity and effector interactions .
Attribute | Value |
---|---|
Gene Symbol | GNG11 |
Chromosome Location | 7q22.1 |
Protein Length | 73 amino acids |
Molecular Weight | ~8.6 kDa (predicted) |
OMIM ID | 604390 |
UniProt Accession | P61952 |
GNG11 is a lipid-anchored, membrane-bound protein. Recombinant forms (e.g., His-tagged) are expressed in E. coli and exhibit >90% purity . Its structure includes a conserved gamma subunit domain critical for G protein beta-gamma dimer formation and signaling .
GNG11 participates in:
G Protein-Coupled Receptor (GPCR) Signaling: Modulates downstream effectors like adenylyl cyclase and phospholipase C .
Reactive Oxygen Species (ROS) Regulation: Overexpression induces ROS generation in specific cell lines (e.g., SUSM-1), linking GNG11 to cellular stress responses .
GNG11 exhibits cytoplasmic and membranous expression across diverse tissues :
Tissue | Expression Level | Subcellular Localization |
---|---|---|
Brain (Cerebral Cortex) | High | Membranous/Cytoplasmic |
Kidney | Moderate | Cytoplasmic |
Liver | Moderate | Cytoplasmic |
Lung | Low | Membranous |
Adipose Tissue | Low | Cytoplasmic |
GNG11 is implicated in lung adenocarcinoma:
Cell Line | GNG11 Effect | Molecular Mechanism |
---|---|---|
SUSM-1 | Growth suppression, ROS induction, abnormal nuclear morphology | Downregulation of antioxidant enzymes (e.g., SOD) |
HeLa | No growth inhibition | Stable antioxidant defense systems |
GNG11 interacts with G protein beta subunits and receptors critical for signal transduction:
GNG11 is a member of the guanine nucleotide-binding protein (G protein) gamma family. It encodes a lipid-anchored, cell membrane protein that functions as part of the heterotrimeric G protein complex involved in transmembrane signaling systems . This protein undergoes carboxyl-terminal processing and participates in G protein-coupled receptor (GPCR) signaling pathways and general signal transduction .
GNG11's primary functions include:
Facilitation of G protein-coupled receptor signaling pathway (GO:7186)
Participation in signal transduction (GO:7165)
GTPase activity (GO:3924)
G-protein beta-subunit binding (GO:31681)
It is primarily localized to the membrane (GO:16020), plasma membrane (GO:5886), and heterotrimeric G-protein complex (GO:5834) .
GNG11 has been identified as a strong regulator of cellular senescence, specifically demonstrating an inductive effect . The primary evidence comes from studies by Hossain et al. (2007), which showed that:
GNG11 induces stress-induced senescence in lung fibroblasts
Both knockout and overexpression studies confirm its regulatory role
GNG11-mediated senescence has been studied in non-cancerous cell lines like TIG-7
The cellular senescence pathway activated by GNG11 appears to be connected to its role in transmembrane signaling and G-protein coupled pathways, though the exact mechanisms require further elucidation. Researchers investigating senescence should consider GNG11 as a potential target for experimental manipulation when studying age-related cellular processes.
The human GNG11 gene:
GNG11 has homologs in several model organisms:
This conservation across species suggests evolutionary importance in fundamental cellular processes. Structurally, GNG11 functions within the heterotrimeric G-protein complex, requiring proper membrane localization for its activity.
Integration analysis of gene expression profiles has identified GNG11 as a hub gene in lung adenocarcinoma . Key findings include:
GNG11 expression is significantly decreased in human adenocarcinoma tissues compared to adjacent normal tissues
This has been verified through both bioinformatic analyses and laboratory qRT-PCR validation
The gene ranks highly in protein-protein interaction networks with a score of 24 in cytohubba analysis
These findings position GNG11 as a potentially important biomarker for adenocarcinoma diagnosis, although its utility for prognosis appears limited based on current data.
GNG11 participates in several important signaling pathways:
G protein-coupled receptor signaling pathway
Core function as part of the heterotrimeric G protein complex
Involved in transducing extracellular signals to intracellular effectors
Chemokine signaling pathway
Module analysis of protein-protein interactions in lung adenocarcinoma identified GNG11 as part of a module significantly enriched in chemokine signaling
This module included 24 nodes with 133 edges, containing genes like NMU, S1PR1, IL6, and CXCL12
The involvement in chemokine signaling suggests a potential role in immune and inflammatory responses in cancer
Connection to other signaling nodes
The dysregulation of these pathways in pathological states like cancer may partially explain the observed downregulation of GNG11 in adenocarcinoma and its identification as a hub gene.
Network analysis has revealed that GNG11 functions alongside several other hub genes in lung adenocarcinoma. The top 10 hub genes identified include:
Gene | Score | Expression in Adenocarcinoma | Function |
---|---|---|---|
GNG11 | 24 | Decreased | G protein subunit, signal transduction |
FPR2 | 23 | Decreased | Formyl peptide receptor, inflammation |
P4HB | 21 | Increased | Protein disulfide isomerase |
PIK3R1 | 20 | Decreased | PI3K regulatory subunit |
CDC20 | 19 | Increased | Cell division cycle protein |
ADCY4 | 18 | Decreased | Adenylate cyclase |
TIMP1 | 17 | Increased | Tissue inhibitor of metalloproteinases |
IL6 | 16 | Decreased | Interleukin, inflammatory cytokine |
CXCL12 | 16 | Decreased | Chemokine |
GAS6 | 16 | Decreased | Growth arrest-specific protein |
These genes collectively form a functional network involved in processes including signal transduction, inflammation, cell cycle regulation, and extracellular matrix remodeling . GNG11's highest score (24) positions it as a central node in this network, suggesting it may have upstream regulatory effects on multiple pathways involved in lung adenocarcinoma.
Based on existing research, effective approaches for studying GNG11 include:
Genetic manipulation techniques:
Cell model selection:
Readouts and assays:
Senescence markers: β-galactosidase activity, cell morphology changes, proliferation arrest
Signaling pathway analysis: Western blotting for downstream effectors of G-protein signaling
Transcriptomic profiling: RNA-seq to identify genes regulated by GNG11
Protein interaction studies:
Co-immunoprecipitation to identify binding partners
Proximity labeling techniques to map the GNG11 interactome in different cellular contexts
When designing experiments, researchers should consider the cell-type specificity of GNG11 function and ensure appropriate controls for both gain and loss of function studies.
For researchers working with large-scale datasets, the following bioinformatic approaches have proven valuable:
Integration of multiple gene expression profiles:
Differential expression analysis:
Functional enrichment analysis:
Gene Ontology (GO) analysis for biological processes, cellular components, and molecular functions
KEGG pathway analysis to identify signaling pathways involving GNG11
Protein-protein interaction (PPI) network construction:
Validation of bioinformatic findings:
qRT-PCR to verify expression differences in clinical samples
Analysis of TCGA database to examine correlations with clinical outcomes
These approaches enable researchers to position GNG11 within broader molecular contexts and generate hypotheses for experimental validation.
Clinical research on GNG11 requires careful methodological planning:
Sample collection and processing:
Expression analysis methods:
Sample size and statistical analysis:
Integration with clinical data:
Correlate GNG11 expression with clinical parameters and outcomes
Consider survival analysis using Kaplan-Meier plots and log-rank tests
Ethical considerations:
Obtain proper informed consent and ethical approval
Comply with institutional and international guidelines for human tissue research
Careful attention to these methodological details will enhance the reliability and translational relevance of GNG11 research in clinical settings.
Several avenues for future research on GNG11 show particular promise:
Mechanistic studies:
Detailed investigation of the molecular mechanisms through which GNG11 induces cellular senescence
Exploration of GNG11's role in the chemokine signaling pathway and its relevance to cancer progression
Characterization of the complete interactome of GNG11 in different cellular contexts
Translational opportunities:
Evaluation of GNG11 as a diagnostic biomarker for lung adenocarcinoma and other cancers
Assessment of its potential as a therapeutic target, particularly in relation to its role in cellular senescence
Development of assays to measure GNG11 activity in clinical samples
Integration with emerging research areas:
Exploration of GNG11's role in the tumor microenvironment and immune response
Investigation of epigenetic regulation of GNG11 expression
Analysis of GNG11 in the context of aging and age-related diseases beyond cancer
Novel experimental approaches:
Application of CRISPR-Cas9 technology for precise genome editing to study GNG11 function
Development of organoid models to study GNG11 in more physiologically relevant systems
Single-cell analyses to understand cell-type specific functions of GNG11
These research directions will advance our understanding of GNG11's fundamental biology and potentially reveal new approaches for diagnosing and treating diseases in which this gene plays a significant role.
The potential contributions of GNG11 research to precision medicine include:
Biomarker development:
Therapeutic targeting:
The role of GNG11 in cellular senescence suggests potential applications in senolytic therapies
Its involvement in multiple signaling pathways provides opportunities for pathway-specific interventions
Modulation of GNG11 activity might sensitize resistant tumors to existing therapies
Patient stratification:
GNG11 expression patterns may identify patient subgroups with different disease mechanisms
This could inform treatment selection and prognostication
Integrating GNG11 status with other molecular and clinical parameters could improve precision
Drug discovery:
GNG11 and its interaction partners represent potential targets for novel therapeutics
High-throughput screening approaches could identify compounds that modulate GNG11 activity
Structure-based drug design targeting GNG11 or its protein interactions
GNG11 is a lipid-anchored, cell membrane protein that forms part of the heterotrimeric G protein complex . This complex consists of alpha, beta, and gamma subunits. The gamma subunit, including GNG11, is essential for the GTPase activity, the replacement of GDP by GTP, and the interaction with G protein effectors .
GNG11 is involved in the G protein-coupled receptor (GPCR) signaling pathway, which is pivotal for various cellular processes . The protein’s beta and gamma chains are necessary for the GTPase activity, which is crucial for signal transduction . This signaling pathway influences many cellular functions, including growth, differentiation, and metabolism.
Human recombinant GNG11 is produced using recombinant DNA technology, which involves inserting the GNG11 gene into a suitable expression system to produce the protein in vitro. This recombinant protein can be used for various research purposes, including studying its role in signal transduction and its potential involvement in diseases.
Research on GNG11 has provided insights into its function and role in cellular signaling. The recombinant form of GNG11 is valuable for studying the protein’s structure, function, and interactions with other molecules. It can also be used in drug discovery and development, particularly in targeting GPCR signaling pathways.