MPP3 (Membrane-Associated Guanylate Kinase, WW and PDZ Domain-Containing Protein 3), also known as DLG3, belongs to the MAGUK (Membrane-Associated Guanylate Kinase) family. These proteins interact with cytoskeletal elements and regulate signaling pathways, cell proliferation, and intracellular junctions . The MPP3 antibody (e.g., catalog number 14650-1-AP) is a research tool designed to detect and study this protein’s expression and function in various biological contexts.
The MPP3 antibody is primarily used to investigate DLG3’s role in cellular processes, including:
Signal transduction: Studying interactions with cytoskeletal proteins and signaling pathways .
Neural development: Analyzing DLG3’s role in maintaining apical junctional complexes and neuronal migration .
Tissue structure: Investigating DLG3’s involvement in retinal pigment epithelium organization and neural retina stratification .
A standardized IHC protocol for the MPP3 antibody is available, though specific steps vary by tissue type and detection system. Key steps include:
Sample preparation: Fixation, embedding, and sectioning of tissue samples.
Antigen retrieval: Heat-based or enzymatic methods to unmask epitopes.
Blocking: Incubation with blocking buffer to reduce nonspecific binding.
Primary antibody incubation: Application of MPP3 antibody (dilution optimized per protocol).
Detection: Use of secondary antibodies conjugated to enzymes (e.g., HRP, AP) or fluorescent tags .
MPP3 refers to myeloid-biased multipotent progenitors, which represent a critical population in the hematopoietic stem cell hierarchy. Recent research has revealed that MPP3 cells are functionally and molecularly heterogeneous, containing a distinct subset of myeloid-primed secretory cells characterized by high endoplasmic reticulum (ER) volume and FcγR expression . These cells serve as a reservoir for rapid production of granulocyte/macrophage progenitors (GMPs) and directly amplify myelopoiesis through inflammation-triggered secretion of cytokines in the bone marrow microenvironment . The significance of MPP3 lies in its newly discovered regulatory function as a self-reinforcing amplification compartment in inflammatory stress and disease conditions, controlling myeloid differentiation through lineage-priming and cytokine production .
MPP3 antibodies are valuable tools for multiple research applications, including:
Western Blotting (WB): For protein expression analysis and quantification of MPP3 in cell lysates
Immunofluorescence (IF): To visualize the cellular localization of MPP3, particularly important for distinguishing ER high and ER low MPP3 subpopulations
Flow Cytometry (FACS): Critical for isolating and characterizing MPP3 populations based on surface markers (e.g., FcγR expression)
Immunohistochemistry (IHC): For tissue section analysis to study MPP3 distribution in different microenvironments
Immunoprecipitation (IP): To study protein-protein interactions involving MPP3
These techniques allow researchers to investigate the multiple roles of MPP3 in hematopoiesis, cellular differentiation, and inflammatory responses.
Selection of an appropriate MPP3 antibody depends on several critical factors:
Target epitope: Determine whether you need an N-terminal or C-terminal targeting antibody based on your research question. N-terminal antibodies (e.g., ABIN2786645) recognize the amino-terminal region of MPP3, which may be particularly useful for detecting full-length protein .
Species reactivity: Verify the antibody's cross-reactivity with your species of interest. Some MPP3 antibodies show broad cross-reactivity across human, mouse, rat, and other species, with predictable reactivity percentages (e.g., 100% for human, mouse, rat; 93% for guinea pig) .
Application compatibility: Confirm the antibody has been validated for your specific application (WB, IF, FACS, etc.) .
Clonality: Consider whether a polyclonal antibody (offering multiple epitope recognition) or monoclonal antibody (offering high specificity) better suits your experimental needs .
Validation data: Review available validation data, including positive controls used (e.g., cell lysates for Western blot) .
For studies focusing on secretory MPP3 populations, antibodies capable of distinguishing FcγR+/ER high MPP3 from FcγR-/ER low MPP3 are particularly valuable .
To properly identify and isolate MPP3 populations, researchers typically use a panel of markers in combination with MPP3 antibodies:
Cell Population | Marker Combination | Function |
---|---|---|
HSCs | Lin−/c-Kit+/Sca-1+/Flk2−/CD48−/CD150+ | Hematopoietic stem cells |
MPP3 | Lin−/c-Kit+/Sca-1+/Flk2−/CD48+/CD150− | Myeloid-biased multipotent progenitors |
MPP4 | Lin−/c-Kit+/Sca-1+/Flk2+ | Lymphoid-biased multipotent progenitors |
GMPs | Lin−/c-Kit+/Sca-1−/FcγR+/CD34+ | Granulocyte/macrophage progenitors |
For specifically identifying the secretory MPP3 subset, additional markers are required:
FcγR expression: Distinguishes myeloid-primed MPP3 subset
ER-Tracker dye or KDEL (ER marker): Identifies ER high MPP3 secretory population
Approximately 31.0 ± 10.4% of the MPP3 compartment is identified as ER high MPP3 at steady state .
Distinguishing between different MPP3 subpopulations requires a multifaceted approach:
Flow cytometry panel design: Combine Lin−/c-Kit+/Sca-1+/Flk2−/CD48+/CD150− markers with FcγR and ER-Tracker dye to separate:
Morphological analysis: Transmission electron microscopy (TEM) can reveal distinct ER structures, with ER high MPP3 showing dense rough ER structures morphologically distinct from GMPs but similar to the secretory apparatus found in specialized immunoglobulin-producing plasma cells .
Immunofluorescence microscopy: Using ER marker KDEL alongside MPP3 antibodies can provide visual confirmation of the ER high subset .
Single-cell RNA sequencing: This approach reveals the molecular heterogeneity of MPP3, allowing identification of distinct clusters representing:
Functional analysis: Secretory assays measuring cytokine production after LPS/Pam3CSK4 stimulation can functionally distinguish MPP3 subsets .
By combining these approaches, researchers can reliably identify and isolate the specific MPP3 subpopulation relevant to their research question.
To effectively study the secretory functions of MPP3, consider these methodological approaches:
Stimulation protocols: Standard protocols use LPS/Pam3CSK4 for inflammatory stimulation before collecting supernatants at 24 hours .
Secretome analysis options:
Key readouts to measure:
Controls: Include HSCs, MPP4, and GMPs as comparative populations to establish MPP3-specific secretory patterns .
Downstream effects assessment: Perform differentiation assays with naïve HSPCs in methylcellulose and liquid culture using supernatants from stimulated versus unstimulated MPP3 .
Note that MPP3 exhibit complex cell type-specific secretory patterns with unique cytokine signatures dependent on stimulation status. Stimulated MPP3 show increased secretion of pro-inflammatory/pro-myeloid differentiation cytokines, while simultaneously modulating regulatory factors .
When encountering inconsistent results with MPP3 antibodies, implement this systematic troubleshooting approach:
Antibody validation:
MPP3 heterogeneity considerations:
Experimental conditions:
Technical optimization:
For Western blotting: Optimize protein extraction methods, blocking conditions, and antibody concentrations
For flow cytometry: Ensure proper compensation and gating strategies
For immunofluorescence: Optimize fixation and permeabilization protocols
Biological variables:
For studying MPP3 in inflammatory conditions, these methodologies have proven particularly effective:
In vivo inflammatory models:
Ex vivo stimulation approaches:
Analytical techniques:
Functional assays:
Time-course experiments:
These approaches allow for comprehensive analysis of how inflammatory signals alter MPP3 function, secretory activity, and contribution to emergency myelopoiesis.
Designing robust experiments to investigate molecular mechanisms of MPP3 function requires a multifaceted approach:
Transcriptional profiling strategies:
Bulk RNA sequencing to compare ER high versus ER low MPP3 populations
Single-cell RNA sequencing to identify distinct molecular clusters
K-means clustering analyses using highly variable genes (HVGs)
Principal component analyses to visualize relationships between MPP3 subsets and other hematopoietic populations (HSCs, GMPs)
Functional genomics approaches:
Protein-level analysis:
Secretory pathway investigation:
Functional validation:
These methodologies collectively enable researchers to dissect the molecular underpinnings of MPP3 function, particularly the distinct roles of ER high/FcγR+ versus ER low/FcγR− subpopulations.
For optimal performance of MPP3 antibodies, follow these storage and handling guidelines:
Storage temperature:
Short-term (≤1 month): 2-8°C
Long-term: -20°C in small aliquots to avoid repeated freeze-thaw cycles
Avoid freezing at -80°C unless specifically recommended by the manufacturer
Working dilutions:
Western blotting: Typically 1:500-1:2000 dilution
Immunofluorescence: Often 1:100-1:500 dilution
Flow cytometry: Usually 1:50-1:200 dilution
Optimization is recommended for each specific application and antibody
Buffer composition:
For dilution: PBS with 0.1% BSA, 0.05% sodium azide, and 0.1% Tween-20
For long-term storage: Add 50% glycerol to prevent freeze-thaw damage
Stability considerations:
Avoid repeated freeze-thaw cycles (create single-use aliquots)
Protect conjugated antibodies from light exposure
Monitor for signs of degradation (loss of specificity, increased background)
Quality control:
Following these guidelines will help maintain antibody integrity and ensure consistent experimental results when working with MPP3 antibodies.
When adopting a new MPP3 antibody for your research, implement these validation methods:
Specificity validation:
Application-specific validation:
For Western blotting: Confirm band appears at expected molecular weight
For flow cytometry: Compare staining pattern with established markers
For immunofluorescence: Verify expected subcellular localization
For immunoprecipitation: Confirm pull-down of known interaction partners
Cross-reactivity assessment:
Orthogonal validation:
Confirm antibody results using alternative detection methods
Compare protein detection with mRNA expression data
Correlation with functional assays specific to MPP3
Reproducibility testing:
Assess lot-to-lot variation if using multiple antibody batches
Test across different sample preparation methods
Verify consistent results across multiple biological replicates
Thorough validation ensures reliable results and prevents misinterpretation of data when studying the complex biology of MPP3 in various experimental contexts.
MPP3 antibodies are instrumental in elucidating hematopoietic stress responses through these research applications:
Identification of stress-responsive populations:
Characterization of emergency myelopoiesis:
Mechanism investigation:
Pathological applications:
Therapeutic target identification:
Using MPP3 antibodies to identify and isolate populations for functional studies
Developing approaches to modulate MPP3 secretory function
Testing interventions that affect the balance between MPP3 subpopulations
MPP3 antibodies thus provide crucial tools for understanding how hematopoietic stem and progenitor cells respond to physiological challenges and contribute to emergency myelopoiesis.
When designing multiparameter flow cytometry panels including MPP3 antibodies, consider these technical aspects:
Panel design strategy:
Fluorophore selection considerations:
Avoid spectral overlap between critical markers
Reserve brightest fluorophores (PE, APC) for key discriminating markers
Consider tandem dyes (PE-Cy7, APC-Cy7) for increased parameters
Controls and compensation:
Single-stained controls for each fluorophore
Fluorescence-minus-one (FMO) controls for accurate gating
Biological controls (known positive and negative populations)
Sample preparation optimization:
Standardize isolation procedures to minimize variability
Use viability dyes to exclude dead cells
Optimize fixation protocols if intracellular staining is required
Critical gating strategy:
Analysis considerations:
Consider dimensionality reduction techniques (tSNE, UMAP) for visualization
Implement consistent batch analysis for longitudinal studies
Correlate flow cytometry findings with functional and molecular data
Proper implementation of these considerations ensures accurate identification and characterization of MPP3 subpopulations in complex experimental settings.
Integrating MPP3 antibody data with single-cell RNA sequencing requires a coordinated analytical approach:
Experimental design integration:
Computational integration approaches:
Analytical strategies:
Use HSC and GMP conserved gene signature lists extracted from independent scRNA-seq analyses
Categorize clusters into functional groups (HSC gene-enriched immature, GMP gene-enriched myeloid-primed, metabolically activated intermediate)
Perform Gene Ontology (GO) and Slingshot analyses for cluster annotation and trajectory prediction
Validation frameworks:
Functional correlation:
Connect transcriptional profiles with secretory capacity
Link gene expression patterns to differentiation potential
Correlate unique cytokine signatures with transcriptional states
This integrated approach provides comprehensive insight into the molecular heterogeneity of MPP3 and connects antibody-defined cellular phenotypes with underlying transcriptional programs.
Several emerging technologies are poised to significantly advance MPP3 antibody applications:
Spatial transcriptomics integration:
Combining MPP3 antibody staining with spatial transcriptomics to map MPP3 subpopulations within bone marrow niches
Correlating spatial distribution with functional heterogeneity
Investigating microenvironmental factors influencing MPP3 differentiation trajectories
Mass cytometry (CyTOF) applications:
Developing MPP3 antibodies conjugated to rare earth metals
Creating high-dimensional panels (30+ parameters) to deeply phenotype MPP3 subsets
Integrating with signaling pathway analysis using phospho-specific antibodies
Live-cell imaging innovations:
Developing non-disruptive MPP3 antibody fragments for live-cell tracking
Implementing intravital microscopy to observe MPP3 behavior in native bone marrow
Tracking cytokine secretion in real-time with reporter systems
Nanobody and single-domain antibody development:
Creating smaller MPP3-targeting antibody fragments for improved tissue penetration
Developing intrabodies for tracking MPP3 in living cells
Engineering bispecific constructs targeting MPP3 and functional markers simultaneously
Multi-omics integration platforms:
Combining antibody-based cell sorting with proteomics, metabolomics, and epigenomics
Implementing CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) to simultaneously capture surface protein and transcript information
Developing computational frameworks for integrating antibody-based data with multiple omics layers
These technological advances will enable researchers to gain unprecedented insight into MPP3 biology, particularly the dynamic changes in MPP3 subpopulations during inflammatory responses and their role in controlling myeloid differentiation.
Despite recent advances, several unresolved questions about MPP3 function could be addressed through innovative antibody-based research:
Heterogeneity beyond current classifications:
Are there additional MPP3 subpopulations beyond the FcγR+/ER high and FcγR−/ER low dichotomy?
Do these populations show tissue-specific variations or disease-specific alterations?
How stable are these phenotypes, and can they interconvert under certain conditions?
Niche interaction dynamics:
How do MPP3 subpopulations interact with specific bone marrow niches?
What cellular and molecular factors regulate the transition from ER low to ER high MPP3?
How does paracrine signaling from MPP3 reshape the bone marrow microenvironment?
Disease relevance:
How are MPP3 populations altered in hematological malignancies?
Could MPP3 secretory dysfunction contribute to inflammatory disorders?
Are there disease-specific MPP3 signatures that could serve as biomarkers?
Developmental origins:
What are the developmental pathways leading to FcγR+/ER high versus FcγR−/ER low MPP3?
How is MPP3 heterogeneity established during ontogeny?
Are there epigenetic mechanisms maintaining MPP3 subpopulation identity?
Therapeutic targeting potential:
Can MPP3 secretory function be selectively modulated for therapeutic benefit?
Would targeting specific MPP3 subpopulations affect emergency myelopoiesis without disrupting homeostatic hematopoiesis?
Could MPP3-directed therapies be developed for inflammatory disorders or malignancies?
Addressing these questions through antibody-based research approaches would significantly advance our understanding of hematopoietic regulation and potentially unveil new therapeutic strategies for various disorders.