PHLDA1 (Pleckstrin Homology-Like Domain Family A Member 1) is a multifunctional protein involved in regulating cell growth, apoptosis, energy homeostasis, and differentiation . The protein contains a pleckstrin homology (PH) domain that is split into N-terminal (β sheets 1–3) and C-terminal (β sheets 4–7 and an α helix) portions .
PHLDA1 appears to play significant roles in:
Regulation of apoptosis, particularly detachment-mediated programmed cell death
Neuronal development-associated apoptosis
Modulation of anti-apoptotic effects of insulin-like growth factor-1 (IGF1)
Translational regulation
Recent research has demonstrated PHLDA1's involvement in various disease processes, including cancer progression, neurological disorders, and inflammatory conditions, making it an important target for investigation .
PHLDA1 antibodies have been validated for multiple research applications:
Western Blot (WB): Detecting PHLDA1 protein expression levels in cell and tissue lysates. Most antibodies can detect the predominant short form of PHLDA1 (~38 kDa) versus the long form (~53 kDa) .
Immunohistochemistry (IHC): Examining PHLDA1 distribution in formalin-fixed, paraffin-embedded tissue sections to evaluate expression patterns in normal versus diseased tissues .
Immunofluorescence (IF): Visualizing PHLDA1 localization within cells using fluorescently labeled secondary antibodies .
Immunocytochemistry (ICC): Detecting PHLDA1 in cultured cells to study subcellular localization .
Co-immunoprecipitation: Isolating PHLDA1 and its binding partners to study protein-protein interactions .
When selecting a PHLDA1 antibody, researchers should verify validation data for their specific application and consider recommended dilutions provided by manufacturers (e.g., 1:2000-1:10000 for ELISA, 1:20-1:200 for IHC) .
PHLDA1 expression varies significantly between normal and diseased tissues, particularly in:
High expression of PHLDA1 positively correlates with survival in MYCN-amplified neuroblastoma patients
PHLDA1 appears to influence neuronal differentiation through interaction with the DCAF7/AUTS2 complex
PHLDA1 is upregulated after subarachnoid hemorrhage (SAH), peaking at 24 hours post-injury
Increased PHLDA1 is predominantly found in microglial cells
PHLDA1 deficiency reduces pro-inflammatory cytokines (IL-1β, IL-6, IL-18) and increases anti-inflammatory IL-10
These expression patterns suggest PHLDA1 may function differently depending on tissue context and disease state, highlighting the importance of experimental controls when using PHLDA1 antibodies .
When performing Western blot analysis with PHLDA1 antibodies, researchers should consider:
The PHLDA1 gene encodes both long and short isoforms
Predicted molecular weights are 45 kDa (long) and 30 kDa (short)
These often appear at 53 kDa and 38 kDa respectively on SDS-PAGE
The short form (~38 kDa) is predominantly expressed in human cells
Effective cell lysis is critical - consider using lysis buffers containing protease inhibitors
For neuronal or brain tissue samples, specialized extraction methods may be needed to overcome high lipid content
Monoclonal antibodies (e.g., EPR6674) offer high specificity and reproducibility
Verify cross-reactivity with target species (most PHLDA1 antibodies are validated for human samples)
Positive controls: U87 and U251 cells show high PHLDA1 expression
Negative/low expression controls: HEB and LN229 cells show relatively low expression
Consider using PHLDA1 knockdown or overexpression samples as specificity controls
HRP-conjugated secondary antibodies with appropriate species reactivity (e.g., Goat anti-Rabbit HRP at 1:2000 dilution)
Enhanced chemiluminescence (ECL) systems provide sensitive detection
These considerations help ensure specific and reproducible detection of PHLDA1 in Western blot experiments .
Researchers have successfully employed several strategies for PHLDA1 knockdown:
siRNA-mediated knockdown:
shRNA-mediated stable knockdown:
Lentiviral or plasmid vectors expressing PHLDA1-targeted shRNA
Allows for stable, long-term knockdown
Example protocol: Transfection of 3 μg shRNA plasmid using JetPRIME reagent, followed by puromycin selection (0.25-0.5 μg/mL)
Established cell lines with stable PHLDA1 knockdown (e.g., S2 and S4 clones) compared to Mock (control shRNA) and WT (non-transduced) cells
Cell density: 1-3 × 10^6 cells per well in 6-well plates
Transfection reagents: JetPRIME, Lipofectamine, or similar reagents
Selection markers: Puromycin resistance for stable selection
Controls: Non-targeting shRNA and GFP-expressing control plasmids to assess transfection efficiency
Western blot analysis to confirm protein reduction
qRT-PCR to verify mRNA knockdown
Functional assays to demonstrate phenotypic effects
In glioblastoma: Slower cell growth, reduced colony formation
In neuroblastoma: Enhanced cellular ATP levels, increased mitochondrial membrane potential, decreased susceptibility to apoptosis
In neuroinflammatory models: Reduced pro-inflammatory cytokines, increased anti-inflammatory IL-10, shifted microglial polarization from M1 to M2 phenotype
These approaches allow researchers to investigate PHLDA1's functions in different cellular contexts and disease models .
PHLDA1 exhibits context-dependent effects on tumor cell growth, with evidence supporting both oncogenic and tumor-suppressive roles:
High PHLDA1 expression positively correlates with survival in MYCN-amplified neuroblastoma patients
PHLDA1 silencing in IMR-32 neuroblastoma cells leads to:
Patient tissue analysis using immunohistochemistry with PHLDA1-specific antibodies
Survival analysis correlating PHLDA1 expression with patient outcomes
Functional studies using gene knockdown and overexpression
Protein interaction studies to identify PHLDA1 binding partners and affected pathways
Phosphoproteome analysis to identify downstream signaling effects
These findings suggest PHLDA1 may function differently depending on tumor type and cellular context, highlighting the need for cancer-specific investigations when targeting PHLDA1 therapeutically .
Optimizing PHLDA1 immunohistochemistry (IHC) across different tissue types requires careful attention to several methodological aspects:
Formalin-fixed, paraffin-embedded (FFPE) tissues are commonly used
Optimal fixation time is critical: overfixation can mask epitopes
Consider antigen retrieval methods appropriate for the specific PHLDA1 epitope
Choose antibodies validated specifically for IHC applications
Consider using monoclonal antibodies for higher specificity
Verify species reactivity (many PHLDA1 antibodies are human-specific)
Brain tissue:
Liver tissue:
Tumor tissues:
Histological score (Hscore) can be used to quantify PHLDA1 levels
Define clear scoring criteria (e.g., median PHLDA1 Hscore as cut-off value for high vs. low expression)
Use digital image analysis software for unbiased quantification
Consider double immunostaining to study co-localization with other markers (e.g., PHLDA1 with microglial markers)
Include positive and negative control tissues with known PHLDA1 expression
For brain tissue, normal brain samples serve as low expression controls
Use isotype control antibodies to assess non-specific binding
These approaches allow for reliable detection and quantification of PHLDA1 across different tissue types in research and potential diagnostic applications .
PHLDA1 has emerged as an important modulator of the NLRP3 inflammasome pathway, particularly in neuroinflammatory conditions:
PHLDA1 blockade inhibits NLRP3 inflammasome signaling in neurological disorders including:
PHLDA1 deficiency reduces inflammatory cytokines (IL-1β, IL-6, IL-18) that are downstream products of inflammasome activation
NLRP3 inflammasome activator (nigericin) reverses the beneficial effects of PHLDA1 blockade, confirming a functional relationship
PHLDA1 appears to regulate microglial polarization through NLRP3 inflammasome signaling
Protein expression analysis:
Functional manipulation:
Microglial polarization assessment:
Cytokine profiling:
In vivo models:
These research approaches have revealed that PHLDA1 blockade ameliorates neuroinflammation by balancing microglial M1/M2 polarization via suppression of NLRP3 inflammasome signaling, suggesting potential therapeutic targets for neuroinflammatory conditions .
PHLDA1 has emerged as a critical regulator of microglial polarization in various neuroinflammatory disorders. Understanding this mechanism requires sophisticated experimental approaches:
PHLDA1 expression increases in activated microglia following neurological injuries such as subarachnoid hemorrhage (SAH) and ischemic stroke
PHLDA1 knockdown shifts microglial polarization from pro-inflammatory M1 phenotype toward anti-inflammatory M2 phenotype
This polarization shift is associated with:
Decreased pro-inflammatory cytokines (IL-1β, IL-6, IL-18)
Increased anti-inflammatory cytokines (IL-10)
Improved neurological outcomes in various disease models
The regulatory effect appears to be mediated through NLRP3 inflammasome signaling
Microglial phenotype characterization:
In vitro modeling:
Primary microglial cultures with PHLDA1 knockdown/overexpression
Microglial cell lines (BV2, HAPI) for mechanistic studies
Co-culture systems with neurons to assess neuroprotective effects
Live-cell imaging to track phenotypic transitions in real-time
Pathway analysis:
PHLDA1 silencing combined with NLRP3 activators (e.g., nigericin)
Phosphoproteomic analysis to identify signaling pathways affected by PHLDA1 manipulation
Protein-protein interaction studies using co-immunoprecipitation and mass spectrometry
Chromatin immunoprecipitation (ChIP) to identify transcriptional targets
Advanced in vivo approaches:
Conditional PHLDA1 knockout specific to microglial cells
Inducible systems to control timing of PHLDA1 manipulation
Intravital microscopy to observe microglial dynamics in living brain tissue
Single-cell RNA sequencing of microglia from different brain regions
Translational relevance assessment:
These approaches reveal that PHLDA1 blockade ameliorates neuroinflammation by balancing microglial M1/M2 polarization via NLRP3 inflammasome suppression, suggesting potential therapeutic targets for conditions including stroke, Parkinson's disease, and subarachnoid hemorrhage .
Investigating PHLDA1 protein interactions through co-immunoprecipitation (co-IP) and mass spectrometry (MS) requires careful experimental design and execution:
Antibody selection:
Lysis buffer considerations:
Pull-down protocol optimization:
Controls and validation:
Sample preparation:
In-gel or in-solution digestion of immunoprecipitated proteins
Peptide fractionation to increase proteome coverage
Consider crosslinking approaches for transient interactions
MS data acquisition strategies:
Data-dependent acquisition for discovery-based approaches
Targeted methods for validation of specific interactions
Quantitative approaches (label-free or isotope labeling) to compare interactomes under different conditions
Data analysis and interpretation:
Filtering against isotype control to remove non-specific binders
Network analysis using platforms like STRING to identify protein complexes
Pathway enrichment analysis using tools like Reactome to identify biological processes
In one study, this approach identified 111 potential PHLDA1-binding partners in neuroblastoma cells
Co-IP-MS identified different PHLDA1 interactors in control vs. antibody-treated neuroblastoma cells
56 proteins were found in both conditions, while 43 new proteins appeared after antibody treatment
Pathway analysis revealed enrichment of specific signaling processes:
Antibody-specific interactors: antimicrobial response and Rho GTPase signaling
Control-specific interactors: glutamate and glutamine metabolism
Shared interactors: protein and RNA metabolism
PHLDA1 interaction with DCAF7 and AUTS2 was confirmed by follow-up Western blot analysis
These methodological considerations enable researchers to reliably identify and characterize PHLDA1 protein interactions, providing insights into its diverse cellular functions and involvement in disease processes .
The literature reveals seemingly contradictory roles for PHLDA1 in cell survival and apoptosis, requiring sophisticated approaches to reconcile these differences:
Pro-survival/oncogenic functions:
Pro-apoptotic/tumor-suppressive functions:
Context-specific expression analysis:
Interactome characterization:
Signaling pathway analysis:
Genetic approaches:
Domain-specific mutations to identify functional regions
CRISPR-Cas9 genetic screens to identify synthetic lethality partners
Inducible expression systems to study temporal effects of PHLDA1
Integrated analysis of clinical data:
Isoform-specific effects: The long (53 kDa) and short (38 kDa) PHLDA1 isoforms may have different or opposing functions
Cellular context dependence: PHLDA1 may interact with tissue-specific factors that determine whether it promotes survival or apoptosis
Pathway interaction model: PHLDA1's role may depend on the status of other signaling pathways (e.g., EGFR, NLRP3 inflammasome)
Threshold effect hypothesis: PHLDA1 may promote survival at moderate levels but trigger apoptosis at very high levels
Temporal dynamics: PHLDA1's function may change depending on exposure time and cellular state
These approaches can help researchers design experiments that account for context-specific functions of PHLDA1 and resolve apparent contradictions in the literature .
Developing isoform-specific antibodies for PHLDA1 presents several technical challenges that require sophisticated approaches:
Isoform characteristics:
Domain structure considerations:
Epitope selection strategies:
Identify unique peptide sequences present in only one isoform
Target splice junction regions where sequences diverge
Consider the three-dimensional structure to identify accessible regions
Use computational epitope prediction tools to identify antigenic regions
Immunization approaches:
Use synthetic peptides representing isoform-unique regions
Develop recombinant protein fragments with isoform-specific regions
Consider multiple host species to overcome tolerance issues
Implement novel immunization protocols (DNA immunization, virus-like particles)
Screening and validation methods:
Multi-step screening against both isoforms to identify differential reactivity
Overexpression systems for each isoform as positive controls
PHLDA1 knockout cells as negative controls
Competitive binding assays with isoform-specific peptides
Validation across multiple applications (WB, IHC, IP) to ensure specificity
Advanced antibody engineering:
Recombinant antibody approaches with affinity maturation
Phage display screening against specific isoforms
Single-cell B cell cloning from immunized animals
Structural biology approaches to guide antibody optimization
Cross-reactivity issues:
Application-specific considerations:
Reproducibility challenges:
These approaches can help researchers develop and validate antibodies capable of distinguishing between PHLDA1 isoforms, enabling more precise investigation of their potentially distinct functions in various cellular contexts .
PHLDA1 appears to significantly impact cellular phosphoproteome networks, with important implications for understanding its diverse functions across different biological contexts:
Neuroblastoma findings:
Glioblastoma research:
Neuroinflammation studies:
Phosphoproteomic mass spectrometry approaches:
Phosphopeptide enrichment strategies:
Titanium dioxide (TiO2) chromatography
Immobilized metal affinity chromatography (IMAC)
Phosphotyrosine-specific antibody enrichment
Quantitative methods:
Label-free quantification
SILAC (Stable Isotope Labeling with Amino acids in Cell culture)
TMT (Tandem Mass Tag) labeling for multiplexed analysis
Data acquisition strategies:
Data-dependent acquisition (DDA)
Data-independent acquisition (DIA)
Parallel reaction monitoring (PRM) for targeted analysis
Kinase activity profiling:
Kinase substrate peptide arrays
Phospho-specific antibody arrays (e.g., RTK arrays used in PHLDA1 studies)
Activity-based protein profiling with ATP probes
Live-cell kinase activity reporters
Computational phosphoproteomics:
Kinase-substrate prediction algorithms
Pathway enrichment analysis
Phosphorylation motif analysis
Integration with protein-protein interaction networks
Temporal modeling of phosphorylation dynamics
Functional validation approaches:
Site-directed mutagenesis of key phosphorylation sites
Pharmacological inhibition of identified kinases
CRISPR-based deletion of phosphorylation sites
Phosphomimetic and phospho-dead mutations to assess functional relevance
Experimental model selection:
Choose cell types where PHLDA1 has established functions (e.g., glioblastoma, neuroblastoma cells)
Consider both PHLDA1 knockdown and overexpression approaches
Include appropriate controls (scrambled siRNA, empty vectors)
Temporal dynamics assessment:
Analyze phosphoproteome changes at multiple time points after PHLDA1 manipulation
Capture both immediate and delayed effects on signaling networks
Consider inducible systems for precise temporal control
Context-dependent analyses:
Compare phosphoproteome effects under different conditions:
Growth factor stimulation
Stress conditions (oxidative stress, nutrient deprivation)
Differentiation signals
Integrate with transcriptomic and proteomic data for systems-level understanding
Translational relevance: