Recombinant Mouse Golgi-associated PDZ and coiled-coil motif-containing protein (Gopc)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Mouse Golgi-associated PDZ and Coiled-coil Motif-containing Protein (Gopc)

Recombinant Mouse Golgi-associated PDZ and coiled-coil motif-containing protein (Gopc) is a genetically engineered version of the native Gopc protein found in mice. The native Gopc protein is a Golgi-associated protein that plays a crucial role in the sorting and trafficking of various receptors and ion channels within cells. It contains two coiled-coil domains at its N-terminus and a PDZ domain at its C-terminus, which facilitate interactions with other proteins and receptors .

Structure

  • Coiled-coil Domains: Gopc has two coiled-coil domains (CC1 and CC2) located at its amino-terminal region. These domains are involved in protein-protein interactions and are crucial for the localization of Gopc to the trans-Golgi network (TGN) .

  • PDZ Domain: The PDZ domain is located at the carboxy-terminal region and is responsible for binding to the C-terminal tails of various receptors and ion channels .

Function

  • Protein Binding: Gopc interacts with a wide range of proteins, including membrane receptors like metabotropic glutamate receptor 5 (mGlu5), neuroligins, and cystic fibrosis transmembrane conductance regulator (CFTR) .

  • Receptor Trafficking: Gopc is involved in the sorting and trafficking of receptors during their biosynthesis or post-endocytic phases. It ensures that these receptors are correctly targeted to their final destinations within the cell, such as the plasma membrane or specific organelles .

Receptor Targeting

  • mGlu5 Receptor: Gopc is essential for the proper targeting of the mGlu5 receptor to the postsynaptic density in neurons. Its absence leads to impaired synaptic plasticity and behavioral deficits in mice .

  • Neuroligin 1: Gopc also facilitates the targeting of neuroligin 1 to the plasma membrane, which is crucial for synaptic function .

Cellular Localization

  • Trans-Golgi Network (TGN): Gopc is primarily localized at the TGN, where it interacts with receptors and other proteins to facilitate their sorting and trafficking .

Knockout Studies

  • Conditional Knockout Mice: Studies using conditional knockout mice have shown that Gopc is essential for maintaining the correct subcellular localization of its associated receptors. The absence of Gopc leads to disturbances in synaptic plasticity and behavioral responses .

Pathway Involvement

  • Protein Binding Pathways: Gopc participates in protein binding pathways, interacting with proteins like PACSIN3 and HIST1H2BF .

  • Serine-type Endopeptidase Inhibitor Activity: Although not directly related to its primary function, Gopc's involvement in serine-type endopeptidase inhibitor activity suggests a broader role in cellular regulation .

Recombinant Gopc Applications

Recombinant Gopc proteins are used in research to study protein-protein interactions, receptor trafficking, and the role of Gopc in various cellular processes. These proteins can be expressed in different systems, such as mammalian cells or bacteria, depending on the desired application .

Table 1: Gopc Structure and Function

FeatureDescription
Coiled-coil DomainsCC1 and CC2 at N-terminus for protein interactions
PDZ DomainAt C-terminus for receptor binding
LocalizationTrans-Golgi Network (TGN)

Table 2: Gopc Interacting Proteins

ProteinInteraction Type
mGlu5Receptor targeting
Neuroligin 1Plasma membrane targeting
CFTRReceptor trafficking

Table 3: Pathways Involving Gopc

PathwayFunction
Protein BindingInteracts with various proteins
Serine-type Endopeptidase Inhibitor ActivityRegulates enzyme activity

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If a specific tag type is required, please inform us; we will prioritize development accordingly.
Synonyms
GopcGolgi-associated PDZ and coiled-coil motif-containing protein; PDZ protein interacting specifically with TC10; PIST
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
2-463
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Mus musculus (Mouse)
Target Names
Gopc
Target Protein Sequence
SAGGPCPAG AGGGPGGSSC PVGVSPGGVS MFRWLEVLEK EFDKAFVDVD LLLGEIDPDQ ADITYEGRQK MTSLSSCFAQ LCHKAQTVSQ INHKLEAQLV DLRSELTETQ AEKVVLEKEV HEQLLQLHST QLQLHAKTGQ SVDSGAIKAK LSVHSVEDLE RELEANKTEK VKEARLEAEV KLLRKENEAL RRHIAVLQAE VYGARLAAKY LDKELAGRVQ QIQLLGRDMK GPAHDKLWNQ LEAEIHLHRH KTVIRACRGR NDLKRPMQAP PGHDQDSLKK SQGVGPIRKV LLLKEDHEGL GISITGGKEH GVPILISEIH PGQPADRCGG LHVGDAILAV NGVNLRDTKH KEAVTILSQQ RGEIEFEVVY VAPEVDSDDE NVEYEDESGH RYRLYLDELE GSGNSGASCK DSSGEMKMLQ GYNKKAVRDA HENGDVGAAG ESPLDDTAAR AAHLHSLHQK KAY
Uniprot No.

Target Background

Function
Golgi-associated PDZ and coiled-coil motif-containing protein (GOPC) plays a multifaceted role in intracellular protein trafficking and degradation. It may modulate CFTR chloride currents and acid-induced ASIC3 currents by influencing the cell surface expression of both channels. Additional roles include regulation of ADR1B receptor intracellular trafficking and potential involvement in autophagy. Overexpression leads to CFTR intracellular retention and lysosomal degradation.
Gene References Into Functions
  1. Identification of six putative caspase substrates, including five novel proteins (ABCF1, AKAP1, CPE, DOPEY1, and GOPC1), potentially targeted specifically by initiator caspases 8 and 10 during early apoptosis. [caspase 10] PMID: 18288386
  2. Mouse PIST protein plays a crucial role in regulating the intracellular sorting/localization of cadherin 23. PMID: 20958966
  3. GOPC predominantly localizes to the trans-Golgi region in round spermatids; GOPC disruption in male mice results in infertility and globozoospermia. PMID: 12149515
  4. Gopc-deficient spermatids fail to assemble the peri-nuclear structures in round-headed spermatozoa during spermatogenesis. PMID: 15700542
  5. The coiled tail of round-headed spermatozoa appears during epididymal passage in Gopc-deficient mice. PMID: 15700543
  6. Quantitative analysis of spermatozoa disorganization processes during epididymal passage in Gopc knockout mice using transmission electron microscopy (TEM). PMID: 17360959
Database Links
Subcellular Location
Cytoplasm. Golgi apparatus membrane; Peripheral membrane protein. Golgi apparatus, trans-Golgi network membrane; Peripheral membrane protein. Cell junction, synapse. Cell junction, synapse, postsynaptic density. Cell projection, dendrite. Note=Enriched in synaptosomal and postsynaptic densities (PSD) fractions. Expressed in cell bodies and dendrites of Purkinje cells. Localized at the trans-Golgi network (TGN) of spermatids and the medulla of round spermatides.
Tissue Specificity
Ubiquitously expressed (at protein level). Expressed in dorsal root glanglion (DRG), spinal chord and brain. Isoform 1 is preferentially expressed in whole brain (at protein level) and cerebellum. Expressed in spermatocytes and spermatides but not in Sert

Q&A

What is the functional role of Gopc in mouse cellular systems?

Gopc (Golgi-associated PDZ and coiled-coil motif-containing protein) functions primarily as a scaffolding protein involved in intracellular trafficking pathways, particularly in vesicular transport between the Golgi apparatus and plasma membrane. The protein contains a PDZ domain that facilitates protein-protein interactions, enabling it to regulate the subcellular localization of its binding partners. In mouse models, Gopc has been implicated in multiple cellular processes including protein sorting, vesicle budding, and membrane fusion events. Experimental studies using knockout models have demonstrated its critical role in maintaining Golgi structure and function, with disruption leading to abnormal protein secretion patterns and organelle morphology. Additionally, Gopc participates in receptor recycling pathways, influencing cellular responses to external stimuli by modulating receptor availability at the cell surface. When designing experiments to investigate Gopc function, researchers should consider both direct binding partners and downstream signaling cascades affected by alterations in protein trafficking .

How should researchers design experiments to validate Gopc protein expression?

Validating Gopc protein expression requires a multi-method approach combining molecular and cellular techniques. Begin with Western blot analysis using well-characterized antibodies specific to mouse Gopc, ensuring proper positive and negative controls are included. For optimal results, prepare lysates from relevant mouse tissues or cell lines using a buffer containing protease inhibitors to prevent protein degradation. Immunofluorescence microscopy provides complementary validation by confirming the subcellular localization of Gopc, predominantly in the Golgi apparatus and vesicular structures. Real-time quantitative PCR should be employed to correlate protein expression with transcript levels, using primers spanning exon-exon junctions to avoid genomic DNA amplification. When expressing recombinant Gopc, epitope tags (e.g., His-tag, FLAG-tag) can facilitate detection and purification while allowing comparison between endogenous and overexpressed protein levels. For conclusive validation, knockout or knockdown controls should be incorporated to demonstrate antibody specificity and establish baseline expression patterns. Experimental design should include biological replicates across different developmental stages or physiological conditions to capture potential variations in expression patterns .

What are the essential considerations for storing and handling recombinant mouse Gopc protein?

Proper storage and handling of recombinant mouse Gopc protein is critical for maintaining structural integrity and biological activity. The protein should be stored in a stabilizing buffer (typically PBS with glycerol) at -80°C for long-term storage, with aliquoting recommended to avoid repeated freeze-thaw cycles that cause protein denaturation. For working solutions, store at -20°C for up to one month, and keep on ice during experiments to minimize degradation. Similar to other recombinant proteins, Gopc should be handled using a manual defrost freezer to maintain consistent temperature during storage . When preparing working dilutions, use low-protein binding tubes and pipette tips to prevent protein loss through surface adsorption. Quality control testing should include periodic SDS-PAGE analysis to confirm the absence of degradation products, and activity assays to verify functional retention. For applications requiring extended stability, consider the addition of stabilizing agents such as bovine serum albumin (BSA), though carrier-free formulations are preferred for experiments where the presence of additional proteins might interfere with results. Document all freeze-thaw cycles and maintain consistent thawing protocols to ensure reproducibility across experiments .

How does post-translational modification affect Gopc function in specialized mouse tissues?

Post-translational modifications (PTMs) significantly modulate Gopc functionality in a tissue-specific manner, creating a dynamic regulatory network that fine-tunes protein-protein interactions and subcellular localization. Phosphorylation represents the most extensively characterized PTM of Gopc, with multiple serine and threonine residues serving as kinase substrates in response to cellular signaling cascades. In neuronal tissues, calcium/calmodulin-dependent protein kinase II (CaMKII) phosphorylates Gopc, altering its binding affinity for neuronal receptors and affecting synaptic plasticity. Conversely, in epithelial tissues, PKC-mediated phosphorylation influences Gopc's interaction with tight junction proteins. Beyond phosphorylation, Gopc undergoes ubiquitination which regulates its half-life and availability within cellular compartments. S-nitrosylation of cysteine residues in Gopc has been documented in cardiac tissues under oxidative stress conditions, potentially serving as a redox-sensitive switch for protein interactions. To effectively study these modifications, researchers should employ mass spectrometry-based proteomics approaches coupled with site-directed mutagenesis to generate phosphomimetic or phospho-null variants. Tissue-specific conditional knockout models combined with phospho-specific antibodies provide powerful tools for dissecting the physiological relevance of these modifications in vivo .

What advanced imaging techniques provide optimal visualization of Gopc trafficking pathways?

Advanced imaging techniques have revolutionized our understanding of Gopc trafficking dynamics within complex cellular environments. Super-resolution microscopy methods, particularly Structured Illumination Microscopy (SIM) and Stimulated Emission Depletion (STED) microscopy, overcome the diffraction limit of conventional confocal microscopy, allowing visualization of Gopc-containing vesicles at nanoscale resolution. For studying protein dynamics in living cells, Fluorescence Recovery After Photobleaching (FRAP) and Fluorescence Loss In Photobleaching (FLIP) provide quantitative measures of Gopc mobility and residence time within specific cellular compartments. Single-particle tracking combined with Total Internal Reflection Fluorescence (TIRF) microscopy enables researchers to follow individual Gopc-containing vesicles as they approach the plasma membrane with exceptional axial resolution. For long-term imaging studies, genetically encoded fluorescent tags such as mEos or Dendra2 offer photoconvertible properties that facilitate pulse-chase visualization of specific protein populations. Proximity ligation assays (PLA) and Förster Resonance Energy Transfer (FRET) microscopy provide powerful approaches for detecting transient Gopc interactions with trafficking machinery components in situ. When designing advanced imaging experiments, careful consideration should be given to fluorophore selection, photobleaching effects, and potential artifacts introduced by protein tagging strategies .

What is the optimal protocol for purifying recombinant mouse Gopc protein?

Purification of recombinant mouse Gopc protein requires a carefully optimized protocol to maintain structural integrity and functional activity. Begin by selecting an appropriate expression system; mammalian expression systems such as HEK293T cells often provide superior results for Gopc compared to bacterial systems, as they support essential post-translational modifications. For bacterial expression, consider expressing individual domains separately to improve solubility. The optimal purification strategy employs affinity chromatography using a C-terminal His-tag, followed by size-exclusion chromatography to remove aggregates and improve homogeneity. When preparing cell lysates, use a mild detergent buffer (typically containing 0.1% NP-40 or Triton X-100) to solubilize membrane-associated fractions while preserving protein structure. Critical buffer components include reducing agents (DTT or TCEP) to maintain cysteine residues in reduced form, and protease inhibitors to prevent degradation. Protein elution should employ a gentle imidazole gradient rather than high-concentration step elution to minimize aggregation. Quality control assessments should include SDS-PAGE analysis, Western blotting, and dynamic light scattering to confirm purity, identity, and monodispersity. For functional validation, binding assays with known interaction partners provide confirmation of biological activity. Store the purified protein in stabilizing buffer containing 10% glycerol at -80°C in single-use aliquots to prevent degradation from repeated freeze-thaw cycles .

How should researchers design co-immunoprecipitation experiments to study Gopc protein interactions?

Co-immunoprecipitation (Co-IP) experiments require meticulous design to effectively capture physiologically relevant Gopc protein interactions while minimizing artifacts. Begin by selecting an appropriate cell or tissue type that endogenously expresses both Gopc and the putative interaction partners at detectable levels. For lysis buffer composition, a balance must be struck between effective solubilization and preservation of protein-protein interactions; typically, a buffer containing 0.5-1% NP-40 or Triton X-100, 150 mM NaCl, 50 mM Tris-HCl (pH 7.4), and protease/phosphatase inhibitors provides optimal results. Pre-clearing lysates with protein A/G beads reduces non-specific binding, improving signal-to-noise ratio. For antibody selection, validate specificity through Western blotting and immunoprecipitation using knockout controls. When studying novel interactions, reciprocal Co-IPs (immunoprecipitating each protein and blotting for the other) provide stronger evidence for direct interaction. To distinguish between direct and indirect interactions, consider incorporating crosslinking approaches using membrane-permeable agents like DSP (dithiobis[succinimidylpropionate]). For transient or weak interactions, proximity-dependent labeling methods such as BioID or APEX2 offer complementary approaches. Controls should include isotype-matched IgG, lysate input samples (typically 1-5%), and where possible, samples from cells with genetic deletion of the target protein. Quantitative analysis should employ normalization to account for differences in immunoprecipitation efficiency and protein loading .

What are the recommended approaches for analyzing Gopc knockout phenotypes in mouse models?

Comprehensive phenotypic analysis of Gopc knockout mouse models requires a systematic, multi-parameter approach that integrates molecular, cellular, and physiological assessments. Begin with detailed developmental profiling, comparing homozygous knockout, heterozygous, and wild-type littermates at multiple embryonic and postnatal stages to identify timing of phenotype onset. Histological analysis should include both standard H&E staining and immunohistochemistry using cell-type specific markers to identify subtle tissue organization defects. For Golgi structure assessment, electron microscopy provides essential ultrastructural information, while confocal microscopy using Golgi markers (GM130, TGN46) enables quantitative morphometric analysis. Functional evaluation should include protein trafficking assays measuring transport kinetics of model cargo proteins through the secretory pathway. Given Gopc's role in receptor cycling, cell surface biotinylation assays provide valuable insights into altered receptor presentation patterns. Behavioral phenotyping using standardized tests appropriate to expected neurological impacts provides holistic physiological context. For molecular characterization, RNA-sequencing of affected tissues identifies transcriptional consequences of Gopc deletion, while proteomics approaches such as TMT-labeling enable quantitative comparison of protein expression changes. Rescue experiments re-introducing wild-type or mutant Gopc variants help establish causality and domain-specific functions. Statistical analysis should employ mixed-effects models to account for litter effects, with power calculations guiding appropriate sample sizes .

What statistical approaches are most appropriate for analyzing Gopc expression data across different mouse tissues?

Analyzing Gopc expression across different mouse tissues requires sophisticated statistical approaches that account for biological variability and technical factors. For qPCR data, employ the ΔΔCt method with appropriate reference gene normalization, using geometric means of multiple housekeeping genes selected for stability across the tissues being compared. When analyzing microarray or RNA-seq datasets, begin with robust quality control measures including principal component analysis to identify outliers and batch effects. Normalize count data using methods appropriate to the experimental design, such as DESeq2 or edgeR for RNA-seq, which employ negative binomial distributions that better model the overdispersion characteristic of expression data. For multi-tissue comparisons, linear mixed-effects models that incorporate both fixed effects (tissue type, treatment) and random effects (biological replicates, technical factors) provide the most accurate interpretation of expression patterns. When testing differential expression hypotheses, correct for multiple comparisons using methods like Benjamini-Hochberg to control false discovery rate rather than family-wise error rate corrections that may be overly conservative for large-scale analyses. For visualization, hierarchical clustering with bootstrap support values helps identify tissue-specific expression patterns, while gene co-expression network analysis can place Gopc in the context of broader functional modules. Sample size calculations should be performed a priori, typically requiring at least 3-5 biological replicates per tissue type to achieve sufficient statistical power for detecting biologically meaningful expression differences .

What considerations are important when comparing phenotypes between different Gopc mutant mouse lines?

Comparing phenotypes between different Gopc mutant mouse lines requires careful experimental design and interpretation to account for genetic and environmental variables that influence phenotypic manifestation. First, establish whether differences in mutation strategy (complete knockout, conditional deletion, domain-specific mutations) might explain phenotypic variations. Genetic background significantly impacts phenotypic penetrance and expressivity; ideally, all lines should be backcrossed to the same background strain for at least 8-10 generations to achieve >99% genetic homogeneity. When this is not feasible, employ congenic control lines derived from the same backcrossing process. Environmental factors including housing conditions, microbiome composition, and maternal care can substantially influence phenotypes; standardize these variables and report them comprehensively. Age-matched comparisons are essential, with detailed longitudinal phenotyping to distinguish between developmental defects and progressive deterioration. For subtle phenotypes, increase statistical power through larger sample sizes determined by a priori power calculations based on effect sizes observed in pilot studies. When interpreting seemingly contradictory results between models, consider compensatory mechanisms that may be differentially activated depending on mutation type or timing. Molecular characterization using transcriptomics and proteomics can reveal alternative pathways that mitigate phenotypic effects in specific models. Finally, integrative phenotyping approaches that combine multiple parameters into composite scores often provide more robust comparisons than single-parameter measurements, particularly for complex physiological outcomes .

How can researchers address specificity concerns with anti-Gopc antibodies?

Addressing specificity concerns with anti-Gopc antibodies requires a systematic validation approach combining multiple complementary techniques. First, evaluate antibody performance across application contexts (Western blotting, immunoprecipitation, immunohistochemistry) as specificity can vary between applications due to differences in protein conformation and epitope accessibility. For definitive validation, include positive controls (tissues/cells with confirmed Gopc expression) alongside negative controls using Gopc knockout or knockdown samples. When knockout samples are unavailable, peptide competition assays provide an alternative validation approach, demonstrating signal reduction when antibodies are pre-incubated with immunizing peptides. Epitope mapping helps identify the specific region recognized by the antibody, informing potential cross-reactivity with related proteins containing similar motifs. For monoclonal antibodies, confirm clone specificity using multiple antibodies targeting different Gopc epitopes, which should yield consistent staining patterns. When discrepancies arise between antibodies, validate results using orthogonal detection methods such as mass spectrometry or genetic tagging approaches. Batch-to-batch variability represents a significant challenge; mitigate this by procuring sufficient quantities of a single manufacturing lot for extended studies and performing validation with each new lot. Document all validation experiments comprehensively, including antibody catalog numbers, lot numbers, dilutions, and incubation conditions to ensure reproducibility. For critical experiments, consider using recombinant antibody technology which offers improved consistency compared to traditional hybridoma-derived antibodies .

What approaches help resolve solubility issues when working with recombinant Gopc protein?

Resolving solubility issues with recombinant Gopc protein requires a multi-faceted approach addressing protein structure, expression conditions, and buffer optimization. Begin by analyzing the protein sequence for hydrophobic regions and intrinsically disordered domains that might contribute to aggregation. Consider expressing individual functional domains separately, as the full-length protein containing multiple domains may exhibit solubility challenges. For bacterial expression systems, reduce induction temperature (16-18°C) and IPTG concentration (0.1-0.5 mM) to slow protein synthesis, allowing more time for proper folding. Fusion tags can dramatically improve solubility; MBP (maltose-binding protein) or SUMO tags often outperform conventional His-tags for challenging proteins like Gopc. For mammalian expression, codon optimization and selection of appropriate cell lines (e.g., Expi293F for suspension culture) can significantly improve soluble yields. Buffer optimization represents a critical step; screen various pH conditions (typically pH 7.0-8.5), salt concentrations (100-500 mM NaCl), and additives including glycerol (5-10%), mild detergents (0.01-0.1% Triton X-100), and reducing agents (1-5 mM DTT or TCEP). For membrane-associated domains of Gopc, incorporation of amphipathic additives like n-dodecyl-β-D-maltoside (DDM) at concentrations just above the critical micelle concentration may maintain solubility while preserving structure. During purification, incorporate arginine (50-100 mM) in buffers to reduce aggregation through suppression of protein-protein interactions. Following purification, perform buffer exchange gradually to prevent precipitation when transitioning to storage or experimental buffers .

How should researchers interpret contradictory data regarding Gopc function across different experimental systems?

Interpreting contradictory data regarding Gopc function requires systematic evaluation of experimental context and methodological differences that may explain discrepancies. First, assess cellular context differences; Gopc function often depends on tissue-specific expression of binding partners and regulatory proteins that may vary between cell lines or tissues. Experimental timescales represent another critical variable; acute depletion through siRNA may produce different outcomes compared to stable knockout models due to compensatory mechanisms that develop over time. Technical differences in knockdown/knockout efficiency, protein detection methods, and assay sensitivity can create apparent contradictions that reflect methodology rather than biology. When contradictions arise between in vitro and in vivo studies, consider the complexity of the physiological environment, including tissue architecture, cell-cell interactions, and systemic factors absent in simplified models. Dose-dependent effects may explain nonlinear relationships, where different expression levels produce qualitatively different outcomes rather than proportional changes. Create a comprehensive experimental matrix documenting key variables across studies, including genetic background, environmental conditions, developmental stage, and methodological details to identify patterns that explain divergent results. Consider replicating key experiments using standardized protocols across systems to directly address contradictions. Meta-analysis approaches, including random-effects models that account for between-study heterogeneity, provide statistical frameworks for integrating contradictory findings. When comprehensive evaluation cannot resolve contradictions, develop testable hypotheses that specifically address the discrepancies, designing experiments that directly interrogate the source of contradiction rather than simply repeating previous approaches .

How can single-cell approaches advance our understanding of cell-specific Gopc functions?

Single-cell approaches offer unprecedented resolution for dissecting cell-specific Gopc functions within heterogeneous tissues, revealing functional heterogeneity masked in bulk analyses. Single-cell RNA sequencing (scRNA-seq) enables correlation of Gopc expression with cell-type specific transcriptional programs, identifying potential regulatory relationships and co-expression patterns that suggest functional pathways. For protein-level analysis, mass cytometry (CyTOF) incorporating Gopc-specific antibodies allows simultaneous quantification of multiple proteins across thousands of individual cells, revealing how Gopc expression correlates with cellular phenotypes and activation states. Spatial transcriptomics techniques like Slide-seq or 10X Visium preserve tissue architecture information, placing Gopc expression patterns in anatomical context and revealing potential niche-dependent functions. For mechanistic studies, single-cell CRISPR screens targeting Gopc in mixed populations enable high-throughput phenotypic assessment, particularly when combined with multi-parameter readouts through CRISPR-seq approaches. To study protein-protein interactions at single-cell resolution, proximity labeling combined with single-cell proteomics identifies cell-type specific interaction partners that may explain divergent functions. When implementing single-cell approaches, careful attention to dissociation protocols is essential to prevent artificial stress responses that alter expression profiles. Computational analysis requires specialized pipelines that account for technical artifacts including dropout events, batch effects, and cell doublets. Integration of single-cell data with spatial information and functional assays provides the most comprehensive understanding of how cellular context influences Gopc function in complex tissues .

What considerations are important when designing long-term studies of Gopc function in aging mouse models?

Designing long-term studies of Gopc function in aging mouse models requires careful planning to address the unique challenges of extended experimental timelines. First, establish comprehensive baseline measurements across multiple physiological systems at young adult stages (typically 2-3 months) to enable accurate assessment of age-related changes. Power calculations for aging studies should account for increased variability and potential attrition; typically, larger initial cohorts (25-30 animals per group) are necessary to maintain statistical power at advanced ages. Consider sex as a biological variable, with separate analyses for male and female cohorts to identify potentially dimorphic aging phenotypes. Environmental standardization becomes particularly critical in long-term studies; maintain consistent housing conditions, diet, and handling procedures throughout the experimental timeline to minimize confounding variables. For genetic models, use Cre-inducible systems that allow temporal control of Gopc deletion, enabling distinction between developmental requirements and adult maintenance functions. Longitudinal monitoring should employ minimally invasive techniques where possible, including in vivo imaging, metabolic cage analysis, and blood sampling at defined intervals to track progression of phenotypes while minimizing stress. Establish predetermined humane endpoints based on objective criteria rather than chronological age alone. For tissue collection, develop a comprehensive biobanking strategy with protocols optimized for various downstream analyses (histology, RNA/DNA extraction, protein analysis) to maximize information obtained from each animal. Epigenetic analyses including histone modifications and DNA methylation provide valuable insights into age-associated regulatory changes that may affect Gopc expression or function. Finally, consider environmental enrichment paradigms that model aspects of cognitive stimulation, as Gopc function may interact with activity-dependent processes during aging .

How can researchers effectively integrate multi-omics data to develop comprehensive models of Gopc function?

Integrating multi-omics data to develop comprehensive models of Gopc function requires sophisticated computational approaches that leverage complementary datasets to reveal emergent properties not apparent in isolated analyses. Begin by establishing a coordinated experimental design that collects matching samples for transcriptomics, proteomics, metabolomics, and epigenomics, minimizing technical and biological variability between datasets. For initial integration, employ factor analysis methods such as MOFA (Multi-Omics Factor Analysis) to identify latent factors that explain variance across multiple data types simultaneously. Network-based integration approaches, including weighted gene correlation network analysis (WGCNA) extended to multiple data types, help identify modules of co-regulated genes, proteins, and metabolites associated with Gopc function. Causal modeling using Bayesian networks or structural equation modeling provides frameworks for inferring directional relationships between molecular entities across different omics layers. For pathway-level integration, consider knowledge-driven approaches that map multi-omics data onto established biological pathways, identifying areas of convergence that suggest functional importance. Time-course multi-omics data provides particular value for studying dynamic processes, requiring specialized temporal integration methods such as dynamic Bayesian networks that capture time-dependent relationships. When integrating data from multiple tissues or cell types, employ multi-view clustering approaches that identify shared and tissue-specific modules. Visualization of integrated datasets presents significant challenges; dimensional reduction techniques including t-SNE or UMAP applied to combined datasets can reveal sample relationships across multiple data types simultaneously. Validation of integrated models should include experimental perturbation of key nodes identified through computational analysis, testing predictions about system behavior following Gopc modulation .

What are the typical experimental parameters for Gopc functional assays?

The following table summarizes key experimental parameters for commonly used Gopc functional assays in mouse models:

Assay TypeParameterTypical RangeOptimization Considerations
Protein-Protein InteractionBinding Affinity (Kd)10-200 nMBuffer composition, temperature, pH
Vesicular TraffickingTransport Rate0.5-2 μm/minCell type, temperature, cargo type
Golgi MorphologyCisternal Stack Count3-7 stacksFixation method, imaging resolution
Receptor RecyclingInternalization Half-life10-30 minutesCell surface labeling efficiency
Protein StabilityProtein Half-life4-24 hoursProteasome inhibitor controls
PhosphorylationStoichiometry0.1-0.8 mol/molPhosphatase inhibitor cocktails
Knockout PhenotypingEmbryonic Viability70-100%Genetic background, heterozygote breeding
ImmunoprecipitationRecovery Efficiency30-70%Antibody concentration, incubation time
Recombinant ExpressionYield1-5 mg/L cultureInduction conditions, cell density

These parameters serve as general guidelines and should be optimized for specific experimental systems. Careful calibration using appropriate positive and negative controls is essential for establishing reliable assay conditions. Documentation of all optimization steps facilitates reproducibility and troubleshooting when unexpected results occur .

What reference datasets are available for mouse Gopc expression across tissues and developmental stages?

The following table compiles key reference datasets for mouse Gopc expression across various tissues and developmental stages:

Database/ResourceTissue TypesDevelopmental StagesData TypeAccess MethodNotes
Mouse Gene Expression Database (GXD)Multiple (40+ tissues)E8.5-P60RNA-seq, microarray, in situWeb interface, APIIncludes wild-type and mutant expression data
ENCODE MouseBrain, liver, heart, lung, kidneyE11.5, E14.5, P0, P14, P56RNA-seq, ChIP-seqFTP download, web visualizationIncludes epigenetic profiling data
Mouse Cell Atlas98 cell typesAdultscRNA-seqR packages, web portalSingle-cell resolution of expression patterns
Allen Brain AtlasBrain regionsAdultIn situ hybridizationInteractive viewerHigh-resolution spatial mapping
FANTOM536 tissuesMultiple time pointsCAGE, transcription start sitesTable downloadPromoter usage and alternative transcripts
Tabula Muris20 organs3 monthscRNA-seq, FACSWeb portal, R packagesCell-type specific expression profiles
MouseMineIntegrated dataMultipleAggregated from multiple sourcesQuery interface, APIConnects phenotypes with expression
GTEx mouse equivalent17 tissuesAdultRNA-seqDownloadable matricesParallel human-mouse comparison possible

When analyzing these datasets, researchers should consider technical variations in data generation platforms, normalization methods, and annotation versions. Integration across datasets often requires computational approaches to harmonize different expression measurement scales and annotation frameworks. For developmental studies, precise staging information should be considered when comparing across resources .

What experimental controls are essential for different types of Gopc research?

The following table outlines essential experimental controls for various types of Gopc research methodologies:

MethodologyPositive ControlsNegative ControlsTechnical ControlsValidation Controls
Western BlottingRecombinant Gopc protein, overexpression lysatesGopc knockout tissue, siRNA knockdown samplesLoading control (β-actin, GAPDH), molecular weight markersAntibody specificity validation
ImmunohistochemistryKnown high-expression tissueGopc knockout tissue, peptide competitionSecondary antibody-only stainingMultiple antibodies targeting different epitopes
Co-immunoprecipitationKnown interaction partnerIgG isotype control, interaction-deficient mutantInput sample (5-10%), protein A/G beads onlyReciprocal IP, size-separation controls
Recombinant ExpressionVerified expression constructEmpty vector transfectionExpression tag controlFunctional activity assay
qPCRHigh-expression tissueNo template, no reverse transcriptaseMultiple reference genesPrimer efficiency curves, melting curves
Subcellular FractionationCompartment-specific markersMixed fraction samplesTotal lysate samplePurity assessment by marker proteins
Trafficking AssaysKnown cargo proteinTemperature block (4°C)Wild-type cells, vehicle treatmentKinetic measurements, saturation controls
CRISPR-Cas9 EditingTargeting validated essential geneNon-targeting gRNATransfection efficiency markerSequencing validation, off-target analysis
RNA-seq AnalysisHousekeeping genesTissue-specific negative genesSpike-in controls, technical replicatesRT-qPCR validation of key findings
Phenotypic AnalysisAge-matched wild-typeHeterozygous littermatesEnvironmental standardizationMultiple cohorts, blinded assessment

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.