plac8.1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the shipping method and destination. For specific delivery estimates, please consult your local distributor.
Target Names
plac8.1
Uniprot No.

Q&A

What is PLAC8/Plac8.1 and what are its main biological functions?

PLAC8 (Placenta-specific 8) is a protein that plays key roles in the regulation of cell proliferation and survival, with significant implications in tumorigenesis and metastasis. It functions as a critical regulator in various cellular processes, particularly those related to cancer development and progression . In cancer cells, elevated PLAC8 expression has been associated with poor prognosis and resistance to therapeutic interventions, positioning it as a potential target for cancer treatment strategies .

In developmental biology, Plac8.1 (the zebrafish ortholog) demonstrates dynamic expression patterns throughout embryogenesis. Initially expressed in a ubiquitous fashion both maternally and zygotically, by 4 days post-fertilization (dpf), Plac8.1 becomes upregulated specifically in gut tissue while being downregulated in other tissues . This shift in expression pattern suggests specialized developmental functions in gut formation and functioning. Furthermore, the intracellular distribution of Plac8.1 changes significantly during development, transitioning from predominantly cytosolic localization at gastrulation onset to becoming progressively enriched at the plasma membrane in later gastrula stages .

In immune contexts, PLAC8 has recently been identified as playing a significant role in modulating monocyte function during sepsis through the ERK signaling pathway. Research has demonstrated that PLAC8 promotes survival, proliferation, and activation of monocytes under septic conditions .

What applications are PLAC8 antibodies validated for in research?

PLAC8 antibodies have been validated for multiple research applications, with specific validation parameters depending on the antibody preparation. Based on available data, most commercial PLAC8 antibodies are validated for:

  • Western Blotting/Immunoblotting: For detection and quantification of PLAC8 protein expression levels in cell and tissue lysates .

  • Immunohistochemistry on Paraffin-embedded tissues (IHC-P): For visualization of PLAC8 localization within tissue sections, including tonsil, spleen, and tumor tissues .

  • Immunofluorescence/Immunocytochemistry (IF/ICC): For examining subcellular localization of PLAC8 in cultured cells, including hepatocellular carcinoma cell lines .

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of PLAC8 in various sample types .

When selecting an antibody for specific applications, researchers should consider the recommended dilutions, which typically vary by application:

  • ELISA: 1:2000-1:10000

  • IHC: 1:20-1:200

  • IF: 1:50-1:200

Validation data, including antibody specificity confirmation through immunoblotting with appropriate controls, is essential before proceeding with experimental applications .

How should researchers interpret PLAC8 expression patterns in normal versus disease states?

Interpreting PLAC8 expression patterns requires careful consideration of cellular context, tissue type, and disease state. In normal physiology, PLAC8 shows tissue-specific expression patterns with developmental regulation, as evidenced by its dynamic expression during zebrafish embryogenesis .

In pathological contexts, particularly cancer, PLAC8 expression often shows significant alterations. For example, in matched primary and metastatic colorectal cancer cell lines, higher PLAC8 levels have been observed in metastatic cells (SW620 vs. SW480 and KM12SM vs. KM12C), suggesting a potential role in metastatic progression . The strongest PLAC8 signals have been detected in microsatellite unstable mucinous cell lines like LoVo .

When analyzing PLAC8 expression in disease states, researchers should:

  • Compare expression levels between normal and pathological tissues from the same origin

  • Assess subcellular localization, as PLAC8 may relocalize from cytosolic to membrane-associated forms during disease progression

  • Correlate expression with clinical parameters, such as tumor stage and patient outcomes

  • Consider post-transcriptional regulation, as PLAC8 protein levels may not always correlate with mRNA expression

In inflammatory conditions like sepsis, PLAC8 has been shown to be highly expressed and correlated with increased levels of inflammatory cytokines (TNF-α, IL-6) and anti-inflammatory cytokines (IL-10) .

How does PLAC8 contribute to epithelial-to-mesenchymal transition (EMT) in cancer progression?

PLAC8 plays a regulatory role in the epithelial-to-mesenchymal transition (EMT), a critical process in cancer progression and metastasis. Research indicates that PLAC8 influences EMT through unconventional mechanisms that affect E-cadherin (CDH1) expression, a key epithelial marker whose repression is characteristic of EMT.

The mechanistic pathway appears to involve ERK signaling. PLAC8 has been shown to promote ERK activation, as evidenced by increased phosphorylation of ERK protein without significant changes in total ERK expression levels . This PLAC8-ERK axis likely contributes to the regulation of cellular phenotypes associated with EMT, including enhanced invasiveness and mobility.

Experimentally, researchers investigating PLAC8's role in EMT should:

  • Assess changes in both mRNA and protein levels of epithelial markers (E-cadherin, ZO-1) and mesenchymal markers (N-cadherin, vimentin)

  • Perform immunofluorescence to examine subcellular localization of these markers

  • Evaluate ERK pathway activation through phospho-ERK analysis

  • Consider cell-autonomous effects through mosaic expression studies

What methodological approaches are recommended for studying PLAC8's role in immune cell function?

Studying PLAC8's role in immune cell function, particularly in contexts like sepsis and cancer immunology, requires a multi-faceted methodological approach:

  • Expression Analysis:

    • Quantitative RT-PCR for mRNA expression of PLAC8 and immune cell markers (CD14, CD16)

    • Western blotting to evaluate protein expression levels and phosphorylation status of downstream targets like ERK1/2

  • Functional Assays:

    • Cell Counting Kit-8 (CCK-8) assay to assess immune cell proliferation in response to PLAC8 modulation

    • Flow cytometry to analyze cell cycle distribution, with particular attention to G0/G1 and S phase populations

    • Cytokine profiling via ELISA to measure secretion of pro-inflammatory (TNF-α, IL-6) and anti-inflammatory (IL-10) cytokines

  • Genetic Manipulation:

    • RNA interference (si-PLAC8) to evaluate loss-of-function effects

    • Overexpression studies using PLAC8-expressing vectors

    • Control experiments with appropriate negative controls (si-plac8-NC, plac8-NC)

  • Pathway Analysis:

    • Examination of ERK pathway activation through phospho-ERK/total ERK ratio analysis

    • Pharmacological inhibition of ERK signaling to confirm pathway dependency

  • In vivo Modeling:

    • Sepsis models (like cecal ligation and puncture) to study PLAC8 regulation in a physiologically relevant context

    • Assessment of immune cell infiltration in tumor models

When designing these experiments, researchers should consider:

  • Appropriate time points for analysis, as PLAC8 effects may vary temporally

  • Cell type-specific responses, as PLAC8 may differentially affect various immune cell subpopulations

  • Proper controls for genetic manipulation and stimulation conditions

  • Combined in vitro and in vivo approaches to validate findings

How can researchers effectively evaluate PLAC8 as a biomarker for immunotherapy response?

Evaluating PLAC8 as an immunotherapy biomarker requires comprehensive assessment across multiple analytical dimensions:

  • Immune Microenvironment Characterization:

    • Apply multiple computational algorithms (TIMER, EPIC, MCPCOUNTER, QUANTISEQ) to quantify immune cell infiltration patterns

    • Analyze correlations between PLAC8 expression and specific immune cell populations (CD8+ T cells, macrophages, NK cells)

    • Evaluate relationships with immune, stromal, and estimate scores to understand the broader immunological context

  • Checkpoint Inhibitor Relationship Assessment:

    • Investigate correlations between PLAC8 and established immune checkpoint molecules

    • Apply the Immunophenoscore (IPS) to evaluate immunotherapeutic potential in patient cohorts

    • Utilize the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm to predict response to immune checkpoint blockade

  • Genomic Integration:

    • Analyze associations with tumor mutation burden (TMB) and microsatellite instability (MSI), established predictors of immunotherapy response

    • Characterize mutational landscapes in tumors with varying PLAC8 expression levels

  • Validation in Clinical Cohorts:

    • Develop stratification approaches based on PLAC8 expression levels

    • Correlate PLAC8 expression with clinical outcomes in immunotherapy-treated patients

    • Consider constructing nomogram models incorporating PLAC8 with other predictive factors

A methodologically sound approach would include:

  • Initial discovery in publicly available datasets (e.g., TCGA)

  • Validation in independent clinical cohorts

  • Functional studies to establish mechanistic links between PLAC8 and immune regulation

  • Prospective evaluation in clinical trials of immunotherapy

Current evidence suggests PLAC8 may be associated with immune dysfunction yet shows complex relationships with the TIDE score and immune exclusion parameters .

What technical considerations are important when using PLAC8 antibodies in developmental research?

When utilizing PLAC8/Plac8.1 antibodies in developmental research, particularly in model organisms like zebrafish, several technical considerations are critical:

  • Antibody Validation for Cross-Species Reactivity:

    • Confirm specificity through immunoblotting with appropriate controls

    • Validate using genetic knockdown/knockout models where possible

    • For zebrafish Plac8.1 studies, researchers have generated specific rabbit polyclonal antibodies against C-terminal peptides

  • Developmental Stage-Specific Protocols:

    • Optimize fixation protocols based on embryonic stage (gastrulation vs. later stages)

    • Adjust permeabilization conditions to account for changing tissue architecture

    • Consider that the subcellular localization of Plac8.1 changes during development (cytosolic at early gastrulation to plasma membrane-enriched at later stages)

  • Phenotype Analysis in Gain/Loss-of-Function Studies:

    • In Plac8.1 overexpression experiments, systematically evaluate dose-dependent effects, as phenotype penetrance increases with concentration

    • Include rescue experiments to confirm specificity (e.g., co-injection with antisense morpholino)

    • Document developmental defects comprehensively (e.g., dorsally curved/shortened body axes, cyclopia)

  • Control for Maternal vs. Zygotic Expression:

    • When studying early development, distinguish between maternally provided and zygotically expressed protein

    • Consider temporal dynamics, as expression patterns change significantly during development (e.g., ubiquitous expression until 4 dpf followed by gut-specific upregulation)

  • Relationship to Conserved Developmental Pathways:

    • Assess effects on canonical developmental markers and processes

    • Evaluate interactions with critical regulatory pathways like E-cadherin regulation

Researchers should note that human PLAC8 RNA injection can produce similar phenotypes to zebrafish Plac8.1 in developmental models, suggesting functional conservation across species .

What are the optimal storage and handling conditions for PLAC8 antibodies?

For optimal performance and longevity of PLAC8 antibodies, researchers should implement the following storage and handling protocols:

  • Storage Buffer Composition:

    • Commercial PLAC8 antibodies are typically supplied in specialized buffers containing:

      • 50% Glycerol

      • 0.01M PBS, pH 7.4

      • 0.03% Proclin 300 as a preservative

    • This formulation helps maintain antibody stability and prevent microbial contamination

  • Physical Storage Conditions:

    • Store antibody aliquots at -20°C for long-term storage

    • Avoid repeated freeze-thaw cycles by preparing appropriate working aliquots

    • For short-term use (within 1-2 weeks), storage at 4°C is acceptable

    • Protect from light, particularly for fluorophore-conjugated antibodies

  • Working Dilution Preparation:

    • Prepare fresh working dilutions on the day of experiment

    • Use high-quality, filtered buffers for dilution

    • For different applications, follow recommended dilution ranges:

      • ELISA: 1:2000-1:10000

      • IHC: 1:20-1:200

      • IF: 1:50-1:200

  • Quality Control Considerations:

    • Include appropriate positive and negative controls in each experiment

    • Periodically validate antibody performance, especially after prolonged storage

    • Document lot-to-lot variations when using commercial antibodies

  • Troubleshooting Guidance:

    • If signal intensity decreases over time, consider preparing fresh antibody dilutions

    • For high background in immunostaining, optimize blocking conditions and increase dilution

    • For weak or absent signals, reduce dilution or extend incubation time

For polyclonal PLAC8 antibodies that have undergone Protein G purification with >95% purity , maintain sterile conditions when handling to preserve specificity and reactivity.

How can researchers differentiate between PLAC8 and its orthologs/paralogs using antibody-based approaches?

Differentiating between PLAC8 and its orthologs/paralogs requires careful selection of reagents and experimental design:

  • Epitope Selection Strategy:

    • Choose antibodies raised against species-specific or divergent regions of PLAC8

    • For zebrafish studies, researchers have successfully generated specific antibodies against C-terminal peptides of Plac8.1

    • Avoid antibodies targeting highly conserved domains if specificity between closely related proteins is required

  • Validation for Cross-Reactivity:

    • Perform systematic validation using:

      • Overexpression systems with tagged variants of each ortholog/paralog

      • Knockdown/knockout models to confirm signal specificity

      • Western blotting to assess reactivity across different species samples

  • Experimental Approaches for Discrimination:

    • Co-immunostaining: Use differentially labeled antibodies against PLAC8 and related proteins

    • Sequential Immunoprecipitation: Deplete one protein first, then assess remaining signal

    • Competitive Blocking: Pre-incubate with recombinant proteins or peptides representing specific orthologs

  • Species-Specific Considerations:

    • For human PLAC8, antibodies like PACO47086 are validated for human samples

    • For studies comparing human PLAC8 and zebrafish Plac8.1, separate validation is essential

    • Note that human PLAC8 RNA injection can produce phenotypes similar to zebrafish Plac8.1 in developmental models, suggesting functional conservation

  • Technical Controls:

    • Include recombinant protein standards representing each ortholog

    • For polyclonal antibodies, consider pre-absorption with related proteins

    • Use genetic models with fluorescent tagging to confirm antibody specificity

A careful experimental design incorporating these approaches can help researchers reliably distinguish between PLAC8 family members in complex biological systems.

What quality control measures should be implemented when using PLAC8 antibodies in quantitative applications?

When employing PLAC8 antibodies for quantitative applications such as protein expression analysis or biomarker assessment, implementing rigorous quality control measures is essential:

  • Antibody Validation Parameters:

    • Specificity Confirmation: Validate using Western blot analysis with appropriate positive and negative controls

    • Sensitivity Assessment: Determine detection limits using standard curves with recombinant proteins

    • Dynamic Range Evaluation: Establish the linear range of detection for quantitative applications

  • Standard Curve Generation:

    • Use purified recombinant human Placenta-specific gene 8 protein (1-110AA) as a reference standard

    • Prepare serial dilutions covering the expected range of endogenous protein

    • Include standards in each experimental run to account for inter-assay variability

  • Internal Controls Implementation:

    • Include housekeeping proteins (like GAPDH, β-actin) for normalization in Western blots

    • For flow cytometry, use isotype controls matched to the PLAC8 antibody

    • Include biological reference samples with known PLAC8 expression levels

  • Assay Validation Metrics:

    • Reproducibility: Assess intra- and inter-assay coefficient of variation (CV)

    • Accuracy: Compare results with orthogonal methods (e.g., mass spectrometry)

    • Robustness: Evaluate performance across different sample types and preparation methods

  • Data Normalization Strategies:

    • For Western blot quantification, normalize to loading controls

    • In cellular assays, consider cell number or total protein normalization

    • For tissue analysis, account for cellular heterogeneity through appropriate reference markers

  • Statistical Considerations:

    • Perform power analysis to determine appropriate sample sizes

    • Apply appropriate statistical tests based on data distribution

    • Include technical replicates (minimum triplicate) for each biological sample

When studying PLAC8's role in pathway activation, such as ERK signaling, always assess both total and phosphorylated forms of downstream targets to calculate activation ratios accurately .

How is PLAC8 being investigated as a therapeutic target in cancer and inflammatory diseases?

PLAC8 is emerging as a promising therapeutic target in both cancer and inflammatory conditions, with several investigational approaches:

  • Cancer Therapeutic Strategies:

    • Targeting EMT Processes: Since PLAC8 promotes EMT through post-transcriptional regulation of E-cadherin , inhibiting this activity could potentially reduce metastatic potential

    • Overcoming Therapy Resistance: PLAC8 expression in cancer cells has been linked to poor prognosis and resistance to therapy , making it a candidate for combination therapy approaches

    • Immune Microenvironment Modulation: PLAC8 influences immune cell infiltration, including CD8+ T cells, macrophages, and NK cells , suggesting potential for enhancing immunotherapy efficacy

  • Inflammatory Disease Applications:

    • Sepsis Intervention: PLAC8 upregulation activates the ERK pathway in sepsis, promoting monocyte survival, proliferation, and activation

    • Cytokine Production Regulation: Modulating PLAC8 affects the production of both pro-inflammatory (TNF-α, IL-6) and anti-inflammatory (IL-10) cytokines

    • Monocyte Function Control: PLAC8 influences CD14 and CD16 expression on monocytes , suggesting potential for targeted intervention in inflammatory disorders

  • Therapeutic Modalities Under Investigation:

    • Small Molecule Inhibitors: Targeting PLAC8 protein-protein interactions

    • RNA Interference: Using siRNA approaches similar to those employed in experimental models

    • Pathway Inhibition: Targeting downstream effectors like ERK in PLAC8-driven pathologies

    • Antibody-Based Therapeutics: Developing neutralizing antibodies against PLAC8

  • Biomarker Integration:

    • PLAC8 expression could potentially stratify patients for personalized therapeutic approaches

    • Combination with other biomarkers might improve prediction of therapy response

  • Challenges in Therapeutic Development:

    • Ensuring target specificity to avoid off-target effects

    • Developing delivery methods for PLAC8-directed therapeutics

    • Balancing PLAC8 inhibition in diseased tissue while preserving normal function

Research in this area remains largely preclinical, with significant opportunities for translational development.

What advanced applications are being developed for PLAC8 antibodies in multiplex imaging systems?

Advanced multiplex imaging applications utilizing PLAC8 antibodies are expanding research capabilities in tumor microenvironment analysis and developmental biology:

  • Multiplex Immunofluorescence and Immunohistochemistry:

    • Co-localization Studies: PLAC8 antibodies can be combined with markers for specific cell populations (immune cells, epithelial cells) to analyze spatial relationships

    • Sequential Multiplex Protocols: Using tyramide signal amplification (TSA) or similar methods to detect multiple targets including PLAC8 on the same tissue section

    • Spectral Unmixing Approaches: Leveraging distinct fluorophores for simultaneous detection of PLAC8 with other proteins of interest

  • Advanced Microscopy Integration:

    • Confocal Microscopy: For high-resolution subcellular localization studies, tracking PLAC8's transition from cytosolic to membrane localization during development

    • Light Sheet Microscopy: For whole-organism imaging in developmental models like zebrafish, where Plac8.1 expression changes dynamically

    • Super-Resolution Techniques: For nanoscale evaluation of PLAC8 interactions with other proteins

  • Spatial Transcriptomics Combination:

    • Integrated Protein-RNA Analysis: Combining PLAC8 antibody detection with in situ hybridization to correlate protein expression with mRNA levels

    • Geographic Information Systems: Mapping PLAC8 expression in relation to tumor regions and microenvironmental features

  • Live Cell Imaging Applications:

    • Antibody Fragment Development: Creating Fab fragments or nanobodies against PLAC8 for live cell imaging

    • Intravital Microscopy: Tracking PLAC8-expressing cells in animal models using fluorescently labeled antibodies

  • Technical Considerations for Multiplex Systems:

    • Antibody Selection: Choose PLAC8 antibodies with minimal cross-reactivity with other targets in the multiplex panel

    • Signal Optimization: Balance signal-to-noise ratios across all channels

    • Spectral Overlap Management: Account for potential bleed-through between fluorophores

    • Image Analysis Pipelines: Develop specialized algorithms for quantifying PLAC8 expression in complex tissue contexts

These advanced applications are particularly valuable for understanding PLAC8's dynamic role in the tumor immune microenvironment, where correlations with various immune cell populations have been observed .

What are common issues encountered when using PLAC8 antibodies and how can they be resolved?

Researchers frequently encounter specific challenges when working with PLAC8 antibodies across various applications. The following troubleshooting guide addresses these issues with practical solutions:

IssuePotential CausesRecommended Solutions
High background in immunostaining- Insufficient blocking
- Too concentrated primary antibody
- Cross-reactivity with related proteins
- Extend blocking time (2-3 hours at RT)
- Increase antibody dilution (try 1:200 for IHC)
- Use species-specific serum in blocking buffer
- Include additional washing steps
Weak or absent signal- Protein degradation
- Insufficient antigen retrieval
- Low PLAC8 expression
- Antibody degradation
- Ensure fresh sample preparation
- Optimize antigen retrieval protocols
- Reduce antibody dilution (try 1:50 for IF)
- Extend primary antibody incubation (overnight at 4°C)
- Use signal amplification systems
Non-specific bands in Western blot- Cross-reactivity
- Sample degradation
- Secondary antibody issues
- Increase antibody dilution
- Use freshly prepared samples with protease inhibitors
- Optimize blocking conditions
- Consider alternative antibody clone
Inconsistent results between experiments- Lot-to-lot antibody variation
- Inconsistent sample preparation
- Protocol variations
- Use consistent antibody lots when possible
- Standardize sample collection and processing
- Document detailed protocols
- Include positive control samples
Discrepancy between protein and mRNA levels- Post-transcriptional regulation
- Protein stability differences
- Technical issues
- Assess both mRNA and protein levels simultaneously
- Consider protein half-life studies
- Optimize extraction protocols for each analyte

When working with developmental systems like zebrafish embryos, additional considerations include:

  • Adjusting fixation protocols based on developmental stage

  • Optimizing permeabilization conditions for embryos at different stages

  • Accounting for the changing subcellular localization of Plac8.1 during development (cytosolic to membrane)

For immune cell applications, ensure proper single-cell isolation techniques and consider the dynamic regulation of PLAC8 during immune activation .

How can researchers overcome challenges in detecting low-abundance PLAC8 in tissue samples?

Detecting low-abundance PLAC8 in tissue samples presents significant technical challenges that can be addressed through specialized methodological approaches:

  • Signal Amplification Strategies:

    • Tyramide Signal Amplification (TSA): Implement this enzyme-mediated amplification system to enhance sensitivity by 10-100 fold over conventional detection

    • Polymer-Based Detection Systems: Utilize multi-polymer conjugates that carry numerous enzyme molecules per antibody binding event

    • Quantum Dots: Consider quantum dot-conjugated secondary antibodies for enhanced signal stability and brightness

  • Sample Preparation Optimization:

    • Preservation Methods: Compare FFPE, frozen sections, and vibratome sections to determine optimal PLAC8 preservation

    • Antigen Retrieval Enhancement: Test multiple retrieval methods (heat-induced vs. enzymatic) and buffers (citrate pH 6.0 vs. EDTA pH 9.0)

    • Section Thickness Adjustment: Use thicker sections (10-15μm) to increase absolute antigen content

  • Antibody Selection and Application:

    • Concentration Optimization: Systematically test a range of dilutions beyond the standard recommendations (1:20-1:200)

    • Incubation Parameters: Extend primary antibody incubation to 48-72 hours at 4°C with gentle agitation

    • Repeated Application: Consider sequential antibody application protocols with intermittent washing

  • Technical Protocol Modifications:

    • Background Reduction: Implement stringent blocking with 5-10% normal serum plus 1% BSA

    • Detergent Optimization: Fine-tune detergent concentration to balance permeabilization with epitope preservation

    • Amplification Controls: Include graduated positive controls to verify detection sensitivity

  • Advanced Detection Approaches:

    • Proximity Ligation Assay (PLA): For detecting PLAC8 interactions with binding partners with single-molecule sensitivity

    • Tissue-CITE-seq: Combine antibody detection with single-cell transcriptomics for multi-omic profiling

    • Mass Cytometry: Consider metal-conjugated antibodies for highly sensitive detection without autofluorescence complications

These approaches are particularly relevant when studying PLAC8 in early disease stages or in tissues where its expression is naturally low but biologically significant.

What emerging technologies are being developed for studying PLAC8 function and regulation?

The field of PLAC8 research is advancing rapidly with several innovative technologies that promise to enhance our understanding of its function and regulation:

  • CRISPR-Based Functional Genomics:

    • CRISPRi/CRISPRa Systems: For precise modulation of PLAC8 expression without genetic deletion

    • CRISPR Screens: To identify synthetic lethal interactions and regulatory networks involving PLAC8

    • Base Editing: For introducing specific point mutations to study structure-function relationships

  • Advanced Protein Interaction Profiling:

    • BioID/TurboID Proximity Labeling: To map the PLAC8 interactome in living cells

    • APEX2-Based Spatial Proteomics: For compartment-specific analysis of PLAC8 interactions

    • Cross-linking Mass Spectrometry: To capture transient PLAC8 protein complexes

  • Single-Cell Multi-Omics:

    • Single-Cell Proteogenomics: Correlating PLAC8 protein levels with transcriptomic changes at single-cell resolution

    • Spatial Transcriptomics: Mapping PLAC8 expression patterns within tissue architecture

    • Cellular Indexing of Transcriptomes and Epitopes (CITE-seq): For simultaneous protein and RNA profiling in immune cell populations

  • Advanced Imaging Technologies:

    • Lattice Light-Sheet Microscopy: For high-speed, low-phototoxicity imaging of PLAC8 dynamics

    • Live-Cell Super-Resolution Imaging: To visualize PLAC8 trafficking and membrane association

    • Correlative Light and Electron Microscopy (CLEM): For ultrastructural localization of PLAC8

  • Translational Research Tools:

    • Patient-Derived Organoids: For studying PLAC8 function in physiologically relevant 3D models

    • Humanized Mouse Models: For investigating PLAC8 function in immune responses within human-like contexts

    • High-Content Drug Screening: To identify compounds modulating PLAC8 activity or expression

These emerging technologies will be particularly valuable for addressing key questions about PLAC8's role in post-transcriptional regulation of E-cadherin , its dynamic subcellular localization during development and disease , and its complex functions in immune cell regulation .

How might PLAC8 research contribute to precision medicine approaches in cancer and inflammatory disorders?

PLAC8 research shows significant potential for advancing precision medicine in both cancer and inflammatory disorders through multiple translational pathways:

  • Biomarker Development for Treatment Stratification:

    • Immunotherapy Response Prediction: PLAC8 expression correlates with immune cell infiltration patterns and may help predict immunotherapy outcomes

    • Chemotherapy Resistance Indicators: PLAC8's association with therapy resistance could guide treatment selection

    • Prognostic Signature Components: Integration of PLAC8 into multi-marker panels for outcome prediction

  • Therapeutic Target Identification:

    • EMT Modulators: Targeting PLAC8's role in post-transcriptional regulation of E-cadherin to prevent metastasis

    • Immune Response Regulators: Modulating PLAC8's effects on monocyte function in inflammatory conditions like sepsis

    • Combination Therapy Approaches: Developing PLAC8 inhibitors to overcome resistance to existing therapies

  • Disease Monitoring Applications:

    • Liquid Biopsy Development: Potential for detecting PLAC8 or PLAC8-expressing cells in circulation

    • Immune Activation Assessment: Monitoring PLAC8 levels in immune cells as markers of inflammatory status

    • Treatment Response Evaluation: Using changes in PLAC8 expression to assess therapeutic efficacy

  • Computational Medicine Approaches:

    • Integration with "-Omics" Data: Incorporating PLAC8 status into multi-parameter models for treatment decisions

    • AI-Based Prediction Tools: Developing algorithms that include PLAC8 expression for outcome prediction

    • Systems Biology Models: Placing PLAC8 within broader pathway networks to identify critical nodes

  • Personalized Cell Therapy Development:

    • Engineered T-Cell Approaches: Potentially targeting PLAC8-expressing malignant cells

    • Monocyte/Macrophage Programming: Modulating PLAC8 to enhance anti-tumor immune responses

    • Stem Cell Differentiation Guidance: Leveraging PLAC8's developmental roles to direct differentiation

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 2025 TheBiotek. All Rights Reserved.