The protein "Recombinant Mouse Uncharacterized protein C10orf118 homolog (Otg1), partial" refers to a specific form of the Otg1 protein, which is a homolog of the human C10orf118 protein . Otg1, or oocyte-testis gene 1, is a murine protein consisting of 890 amino acids . The "recombinant" designation indicates that this protein is produced using recombinant DNA technology, involving the insertion of the Otg1 gene into a host cell for expression . The term "partial" suggests that the recombinant protein may not represent the full-length Otg1 protein but rather a fragment or portion of it .
C10orf118, also known as coiled-coil domain-containing protein 186 or Q7z3E2, is a protein that, in humans, is also called golgin104 . Golgins participate in vesicle tethering and transport . C10orf118 appears to regulate the maturation of dense core vesicles and control the post-Golgi retention of cargos in neurons . The expression of C10orf118 is associated with a higher level of expression of the estrogen receptor, which correlates with positive outcomes in cancer .
C11orf96 is rich in Ser and has multiple predicted phosphorylation sites . Protein interaction prediction analysis revealed that the protein is associated with several transmembrane family proteins and zinc finger proteins . C11orf96 is distributed in all tissues and organs, with the highest expression levels in the kidney, suggesting that it may play a specific biological role in the kidney .
C10orf118 induces hyaluronan (HA) secretion by up-regulating the HAS2 gene in fibroblasts and has been identified in breast cancer tissue specimens . Silencing C10orf118 reduces the mRNA levels of HAS2 and its epigenetic regulator HAS2-AS1, while increasing the levels of the HA receptor CD44 . C10orf118 influences HA metabolism .
C10orf118 is highly expressed by breast tumor cells and is associated with low aggressiveness of cancer . High expression of C10orf118 is positively correlated to patient survival and to a low metastasis .
| Protein | Expression in ZIP8-KO Cells |
|---|---|
| C1orf198 | Downregulated |
| C9orf85 | Downregulated |
| C17orf75 | Downregulated |
| CXorf38 | Downregulated |
Otg1 (oocyte testis gene 1) encodes a Golgi-localized protein with a broad tissue distribution in mice. The full-length transcript has 16 exons that encode a 917-amino acid peptide. The OTG1 protein contains several coiled-coil domains that occupy almost half of the peptide sequence. Confocal microscopy studies confirm its predominant localization in the Golgi apparatus with a Pearson's Correlation of 0.7011 compared to 0.3804 in the endoplasmic reticulum . Although the primary sequence contains a KDEL signal at position 677-680 (compatible with ER retention), it shows preferential localization to the Golgi apparatus.
Current research indicates that Otg1/C10orf118 functions as:
A regulator of hyaluronan synthesis by acting on the up-regulation of the HAS2 gene in fibroblasts
A potential controller of hormone secretion affecting glucose homeostasis
The protein structure contains coiled-coil regions reminiscent of golgin coiled-coil proteins, which are known to function as membrane and cytoskeleton tethers. Although it lacks a transmembrane or small GTPase interacting signal typically found in golgins, OTG1 may still participate in similar intracellular activities by serving as a molecular partner to typical golgins .
Otg1 disruption in mice leads to several significant phenotypes:
| Phenotype | Observation | Comparison to Wild-type |
|---|---|---|
| Postnatal survival | 46.5% die within P1, others gradually die within 30 days | 96.4% wild-type survive past one month |
| Growth | Severe growth retardation | Body weight increase much slower than controls |
| Blood glucose | Decreased to ~25% below normal within two days of birth | Normal levels in wild-type littermates |
| Serum insulin | 0.02 ng/ml in P11 homozygotes | 0.66 ng/ml in wild-type littermates |
| Serum leptin | Below detectable level (<0.2 ng/ml) | Detectable in controls |
| Serum growth hormone | 35% reduction | Normal levels in controls |
| Hepatic IGF-1 expression | 50% decrease at P1, 87% decrease at P11 | Normal expression in controls |
Notably, Otg1 mutant mice exhibit lipohypotrophy and typically die before reaching 5 grams in body weight .
Otg1 appears to play a critical role in the vesicle trafficking machinery of the Golgi apparatus. Studies of Otg1-deficient mice have revealed:
Reduced levels of circulating hormones including insulin, leptin, and growth hormone
Decreased hepatic IGF-1 expression
Impaired glucose homeostasis
The mechanistic link involves OTG1's coiled-coil domains, which are structurally reminiscent of golgin proteins known to function as tethers in vesicle trafficking. Although OTG1 lacks the transmembrane or GTPase-interacting signals typical of golgins, it likely functions as a molecular partner to these proteins, contributing to vesicle capture and providing specificity to the tethering step. When this function is disrupted, the secretion of multiple hormones is affected, leading to the observed metabolic phenotypes .
Research has revealed an interesting correlation between C10orf118 expression levels and breast cancer aggressiveness:
Expression analysis across different breast cancer cell lines showed that C10orf118 gene expression was more pronounced in breast cancer cells compared to stromal cells
The highest expression was found in less aggressive breast cancer cell lines (8701-BC and MCF-7)
Lower expression was observed in the highly aggressive MDA-MB-231 cell line
High expression of C10orf118 is positively correlated with patient survival and low metastasis
C10orf118 expression was associated with the presence of estrogen receptor, which characterizes a good patient survival outcome
These findings suggest that C10orf118 may function as a tumor suppressor in breast cancer, with its expression potentially serving as a prognostic indicator.
C10orf118 has been identified as a novel regulator of hyaluronan (HA) synthesis in the tumor microenvironment. Experimental evidence indicates:
Secreted C10orf118 induces hyaluronan synthase 2 (HAS2) expression in fibroblasts
Knockdown of C10orf118 in MCF-7 cells reduced the mRNA levels of HAS2 and its epigenetic regulator HAS2-AS1
C10orf118 silencing increased levels of the HA receptor CD44
These findings suggest an autocrine effect of C10orf118 on key enzymes and receptors related to HA metabolism
This regulatory role has significant implications for understanding tumor-stromal interactions, as alterations in HA synthesis and size affect tumor growth and metastasis .
When investigating Otg1/C10orf118 function in vitro, consider the following experimental approaches:
Gene silencing by siRNA:
Subcellular localization studies:
Vesicle trafficking assays:
Hormone secretion measurements:
For analyzing the effects of Otg1/C10orf118 on gene expression, implement the following methods:
Quantitative RT-PCR:
RNA-Seq analysis:
Compare transcriptomes of control and Otg1/C10orf118-silenced cells
Analyze differentially expressed genes using DESeq2 or edgeR
Perform pathway enrichment analysis to identify affected biological processes
Validate key findings with qRT-PCR
Chromatin immunoprecipitation (ChIP):
Investigate potential interactions between C10orf118 and chromatin
Use antibodies against C10orf118 or associated transcription factors
Analyze enrichment at promoters of regulated genes (e.g., HAS2)
Promoter reporter assays:
Clone promoter regions of target genes into luciferase reporter constructs
Co-transfect with C10orf118 expression vectors or siRNAs
Measure promoter activity to assess direct transcriptional effects
When designing experiments to study multiple factors affecting Otg1 function, fractional factorial designs can be particularly useful:
Identify key factors for investigation:
Cell type (e.g., fibroblasts, epithelial cells, cancer cell lines)
Growth conditions (serum concentration, hypoxia)
Protein expression levels (overexpression, knockdown)
Stimuli (growth factors, hormones)
Choose appropriate fractional factorial design:
Example design for a 4-factor experiment:
| Run | Cell Type | Oxygen Levels | Otg1 Expression | Growth Factor |
|---|---|---|---|---|
| 1 | Fibroblast | Normoxia | Normal | Present |
| 2 | Fibroblast | Hypoxia | Knockdown | Absent |
| 3 | Cancer | Normoxia | Knockdown | Absent |
| 4 | Cancer | Hypoxia | Normal | Present |
| 5 | Fibroblast | Normoxia | Knockdown | Present |
| 6 | Fibroblast | Hypoxia | Normal | Absent |
| 7 | Cancer | Normoxia | Normal | Absent |
| 8 | Cancer | Hypoxia | Knockdown | Present |
Analysis considerations:
This approach allows for efficient screening of multiple factors affecting Otg1 function while minimizing the number of experimental runs required.
When studying the effects of recombinant Otg1 on cellular functions, the following controls are essential:
Expression controls:
Empty vector controls for overexpression studies
Non-targeting siRNA/shRNA for knockdown experiments
Western blot verification of protein expression levels
Immunofluorescence to confirm subcellular localization
Functional controls:
Known Golgi-disrupting agents (e.g., Brefeldin A) as positive controls
Trafficking assays with well-characterized cargo molecules (e.g., VSVG-GFP)
Measurement of secreted proteins unrelated to Otg1 pathways
Cell-type controls:
Compare effects across multiple cell types (e.g., fibroblasts, epithelial cells)
Use cells with naturally occurring high and low Otg1 expression
Include isogenic cell lines differing only in Otg1 expression
Rescue experiments:
Re-expression of wild-type Otg1 in knockout/knockdown cells
Domain-specific mutants to identify functional regions
Chimeric proteins to assess domain-specific functions
Time-course controls:
Monitor effects at multiple time points after manipulation
Include pre-treatment measurements as baselines
Consider both acute and chronic effects of Otg1 manipulation
When addressing contradictory findings regarding Otg1/C10orf118 function across different cell types:
Systematic comparison approach:
Create a standardized experimental framework to test Otg1 function across cell types
Control for variables such as expression level, culture conditions, and passage number
Quantify function using identical readouts (e.g., same trafficking assay)
Cell type-specific factor analysis:
Investigate expression levels of potential Otg1 interaction partners in each cell type
Assess differences in post-translational modifications of Otg1
Compare subcellular localization patterns using co-localization coefficients
Integrative data analysis:
Perform meta-analysis of results across studies
Use statistical methods that account for between-study heterogeneity
Identify consistent vs. variable aspects of Otg1 function
Reconciliation framework:
Develop models that incorporate cell type-specific factors
Test hypotheses that could explain divergent results (e.g., cell type-specific binding partners)
Design experiments specifically to distinguish between competing models
For example, the apparent contradictions between Otg1's roles in vesicle trafficking and hyaluronan synthesis may be reconciled by understanding how these pathways interact in different cellular contexts. The differential expression of C10orf118 in aggressive vs. less aggressive cancer cell lines suggests context-specific functions that may explain seemingly contradictory observations .
For analyzing Otg1 knockout phenotypes in mouse models, implement these statistical approaches:
Survival analysis:
Kaplan-Meier curves to compare survival between genotypes
Log-rank test to assess statistical significance
Cox proportional hazards model to account for covariates
Example: In Otg1 studies, 46.5% homozygous PB/PB mutants died within P1, while 96.4% wild-type and 94.2% heterozygous littermates survived beyond one month
Longitudinal growth analysis:
Metabolic parameter analysis:
ANOVA with post-hoc tests for comparing multiple groups
Non-parametric alternatives (e.g., Kruskal-Wallis) for non-normally distributed data
Repeated measures designs for glucose tolerance tests
Example: Blood glucose levels in PB/PB Otg1 mice dropped to approximately 25% below normal within two days of birth
Gene expression analysis:
Sample size considerations:
Account for potential early lethality when designing studies
Include heterozygotes to assess potential gene dosage effects
Match for sex, age, and litter when possible
These approaches enable robust analysis of complex phenotypes while accounting for the unique challenges of working with models that exhibit significant survival and developmental effects.
To identify Otg1/C10orf118 interaction partners and signaling pathways, researchers should consider:
Proximity-dependent labeling approaches:
BioID or TurboID fusion proteins to identify proximal proteins in living cells
APEX2 proximity labeling for temporal resolution of interactions
Comparative analysis across cell types and conditions
Co-immunoprecipitation coupled with mass spectrometry:
Both endogenous and epitope-tagged approaches
Crosslinking strategies to capture transient interactions
Quantitative proteomics to assess interaction dynamics
Functional genomics screens:
CRISPR-Cas9 screens to identify genes that modify Otg1 phenotypes
Synthetic lethal screens in Otg1-deficient backgrounds
Targeted screens focused on vesicle trafficking and Golgi function genes
Structural biology approaches:
Domain-specific interaction mapping
Cryo-EM analysis of Otg1 in complex with binding partners
Structure-function studies with domain deletion mutants
Signaling pathway analysis:
Phosphoproteomic analysis in response to Otg1 manipulation
Transcriptional profiling to identify downstream effectors
Small molecule inhibitor screens to disrupt Otg1-dependent pathways
These approaches would provide crucial insights into how Otg1/C10orf118 interfaces with cellular machinery to regulate vesicle trafficking, hormone secretion, and hyaluronan synthesis.
To fully understand the physiological significance of Otg1/C10orf118, researchers should implement these interdisciplinary approaches:
Systems biology integration:
Multi-omics data integration (transcriptomics, proteomics, metabolomics)
Network analysis to position Otg1 within cellular pathways
Mathematical modeling of vesicle trafficking and secretion dynamics
Translational research connections:
Correlation of Otg1 expression with disease states in patient samples
Analysis of human genetic variants in C10orf118 and their phenotypic associations
Development of biomarkers based on Otg1 function or expression
Advanced imaging technologies:
Super-resolution microscopy to visualize Otg1-dependent trafficking events
Live-cell imaging with optogenetic control of Otg1 function
Correlative light and electron microscopy to link Otg1 localization with ultrastructural features
Tissue engineering approaches:
3D organoid models to study Otg1 in physiologically relevant contexts
Co-culture systems to investigate cell-cell communication involving Otg1
Microfluidic platforms to assess dynamic secretory processes
Collaborations across disciplines:
Developmental biologists to study Otg1's role in embryogenesis
Endocrinologists to investigate hormone secretion defects
Cancer biologists to explore tumor-suppressive mechanisms
Glycobiologists to examine hyaluronan metabolism
These interdisciplinary approaches would provide a comprehensive understanding of Otg1/C10orf118's diverse functions and potential therapeutic relevance across multiple biological contexts and disease states.