PLPPR1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

The PLPPR1 antibody is a highly specific immunological reagent designed to target the Phospholipid Phosphatase-Related Protein Type 1 (PLPPR1), a six-transmembrane protein enriched in the brain. PLPPR1 is implicated in modulating cytoskeletal dynamics, cell migration, and axon regeneration by interacting with RhoGDI to regulate RhoA and Rac1 GTPase activity . The antibody is used in research to study PLPPR1’s role in neurobiology, cancer, and developmental biology.

Development and Production

Immunogen: The PLPPR1 antibody (HPA014968) is produced using recombinant human PLPPR1 protein fragments, ensuring high specificity .
Production: Affinity-purified via Protein A chromatography, the antibody is validated through stringent protocols, including immunohistochemistry (IHC) on 44 normal human tissues and 20 cancer types .

Applications

  • Immunohistochemistry: Detects PLPPR1 in tissue sections, revealing expression in brain, kidney, and pancreas .

  • Immunofluorescence: Maps subcellular localization (plasma membrane and cytoplasm) in the Human Cell Atlas project .

  • Western Blot: Validates protein expression in cell lysates of Neuro-2a cells overexpressing PLPPR1 .

Validation and Specificity

Validation MethodKey Findings
IHC on Tissue ArraysStrong staining in brain (cerebellum, cortex) and weak in liver .
Protein ArrayNo cross-reactivity with 363 other human proteins .
Western BlotDetects a ~40 kDa band in lysates of PLPPR1-transfected cells .

Research Findings

Neurological Studies:

  • PLPPR1 overexpression rescues neurons from CSPG-induced axon collapse via RhoA/Rac1 modulation .

  • Antibody staining confirms PLPPR1 localization at sites of axon regeneration in spinal cord injury models .

Cancer Biology:

  • PLPPR1 expression correlates with reduced migration in neuroblastoma cells, suggesting a role in metastasis suppression .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically ship PLPPR1 Antibody orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery timelines.
Synonyms
PLPPR1; LPPR1; PRG3; Phospholipid phosphatase-related protein type 1; Lipid phosphate phosphatase-related protein type 1; Plasticity-related gene 3 protein; PRG-3
Target Names
PLPPR1
Uniprot No.

Target Background

Database Links

HGNC: 25993

KEGG: hsa:54886

UniGene: Hs.382683

Protein Families
PA-phosphatase related phosphoesterase family
Subcellular Location
Membrane; Multi-pass membrane protein.

Q&A

What is PLPPR1 and why is it significant in neuroscience research?

PLPPR1 (Phospholipid Phosphatase-Related Protein Type 1) is a member of the lipid phosphatase-related protein family characterized by six transmembrane domains. It is highly enriched in the brain and has demonstrated pleiotropic functions in several different cellular contexts. The significance of PLPPR1 in neuroscience stems from its ability to induce membrane protrusions, reduce cell motility, increase cell adhesion to extracellular matrix substrates, and overcome inhibitory factors that typically prevent neurite outgrowth . These properties make PLPPR1 an important target for studies on neural development, axon regeneration, and central nervous system injury. Recent research has shown that PLPPR1 can promote axon regeneration after spinal cord injury in mice, highlighting its potential therapeutic relevance in neurological disorders .

How can I validate the specificity of a PLPPR1 antibody?

Validating the specificity of a PLPPR1 antibody requires multiple complementary approaches:

  • Positive and negative controls: Test the antibody in cells with confirmed PLPPR1 expression (such as brain tissue) versus cells with no detectable PLPPR1 expression (such as Neuro2a cells, which do not express detectable levels of PLPPR1 mRNA according to data from NCBI GEO database) .

  • siRNA/shRNA knockdown: Reduce endogenous PLPPR1 expression through RNA interference and confirm reduced signal with your antibody.

  • Overexpression systems: Compare signal in cells transfected with EGFP-PLPPR1 versus EGFP alone as demonstrated in multiple studies .

  • Western blot analysis: Verify that the antibody detects a band of the appropriate molecular weight (~47 kDa for PLPPR1).

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide prior to application to verify signal elimination.

  • Cross-reactivity assessment: Test for cross-reactivity with other PLPPR family members by expressing each individually.

What are the typical expression patterns of PLPPR1 in neural tissues?

PLPPR1 shows enriched expression in the brain compared to other tissues . Within neural tissues, PLPPR1 expression can be detected using real-time PCR from brain tissue preparations, as demonstrated with RNA prepared from P1 brain of C57BL/6J mice . The protein appears to be developmentally regulated, with expression patterns changing during neural development. When designing experiments to detect PLPPR1, it is important to note that some cell lines commonly used in neuroscience research, such as Neuro2a cells, do not express detectable levels of PLPPR1 mRNA under normal conditions . This makes these cell lines useful negative controls or suitable models for exogenous expression studies.

What mechanisms underlie PLPPR1's modulation of RhoA and Rac1 signaling?

PLPPR1 modulates RhoA and Rac1 activation through a novel mechanism involving its association with RhoGDI1. This interaction affects how these small GTPases respond to stimuli such as lysophosphatidic acid (LPA). The mechanistic pathway involves:

This mechanism explains how PLPPR1 can influence cellular processes dependent on RhoA/Rac1 signaling, including cytoskeletal dynamics, cell adhesion, and neurite outgrowth.

How can I design experiments to study PLPPR1's effects on cell adhesion and migration?

To effectively study PLPPR1's effects on cell adhesion and migration, consider the following experimental approaches:

Cell Adhesion Experiments:

  • Attachment assay: Transfect cells with EGFP-PLPPR1 or EGFP control, plate on fibronectin substrate for one hour, then subject to media washes every 15 minutes. Count attached cells at each time point .

  • Detachment assay: Allow PLPPR1-expressing and control cells to attach overnight, then measure their resistance to detachment by trypsin/EDTA treatment .

  • Quantification methods: Use either manual counting of cells in multiple fields or fluorescence-based quantification if using fluorescently tagged proteins.

Cell Migration Experiments:

  • Live cell imaging: Track individual cell trajectories over time (6-12 hours) using time-lapse microscopy as described in previous studies .

  • Analytical parameters: Measure velocity, total distance traveled, and directional persistence.

  • Substrate considerations: Use uniform fibronectin coating (10 μg/ml) to provide a consistent migration environment .

ParameterControl (EGFP)PLPPR1-expressing cellsExpected difference
Adhesion rateLowerHigher↑ ~30-50%
Detachment resistanceLowerHigher↑ ~25-40%
Migration velocityHigherLower↓ ~40-60%
Total distance traveledHigherLower↓ ~30-50%

What methodologies are recommended for analyzing PLPPR1's impact on neurite outgrowth?

To analyze PLPPR1's impact on neurite outgrowth, especially in inhibitory environments, implement the following methodologies:

  • Preparation of inhibitory substrates: Coat glass coverslips with poly-L-lysine (PLL) alone or PLL plus chondroitin sulfate proteoglycans (CSPGs) to create control and inhibitory growth conditions .

  • Transfection of neurons: Use calcium phosphate or lipid-based transfection methods to express EGFP-PLPPR1 or EGFP control in primary hippocampal neurons .

  • Neurite measurements:

    • Total neurite length per neuron

    • Length of the longest neurite

    • Number of primary neurites

    • Branching complexity (Sholl analysis)

  • LPA challenge experiments: Treat neurons with lysophosphatidic acid (4-16 μM) to induce neurite retraction, then compare the responses between PLPPR1-expressing and control neurons .

  • Immunostaining: Use appropriate cytoskeletal markers (e.g., βIII-tubulin, F-actin) to visualize neurite morphology.

  • Quantitative analysis: Employ automated image analysis software (e.g., ImageJ with NeuronJ plugin) for unbiased quantification.

  • Statistical analysis: Use unpaired Student's t-tests or ANOVA with appropriate post-hoc tests (e.g., Tukey's multiple comparison test) to determine significance .

What are the optimal conditions for immunoprecipitation of PLPPR1 and its binding partners?

For successful immunoprecipitation of PLPPR1 and its binding partners:

  • Lysis buffer selection: Use a buffer that preserves protein-protein interactions while effectively solubilizing membrane proteins. A recommended formulation includes:

    • 50 mM Tris-HCl, pH 7.5

    • 150 mM NaCl

    • 1% NP-40 or 1% Triton X-100

    • 0.5% sodium deoxycholate

    • Protease inhibitor cocktail

    • Phosphatase inhibitor cocktail

  • IP strategy for PLPPR1-GFP fusion proteins:

    • Use GFP magnetic/agarose beads as successfully employed in previous studies

    • Incubate cell lysates (600 μg protein) with beads at 4°C for 1-2 hours

    • Perform thorough washes (at least 3-5 times) with lysis buffer

  • RhoGDI co-immunoprecipitation:

    • Immunoprecipitate RhoGDI using specific antibodies

    • When analyzing interactions following LPA treatment, maintain cells in serum-starved conditions (16-18 hours) before treatment

    • Apply LPA at concentrations between 4-16 μM for 2 minutes before lysis

  • Detection strategies:

    • For immunoblotting, transfer proteins to PVDF membranes

    • Block membranes in 5% nonfat dried milk in either PBST or TBST

    • Incubate with primary antibodies overnight at 4°C

    • Detect using sensitive chemiluminescent substrates such as KPL LumiGlo

How can I optimize western blotting protocols for PLPPR1 detection?

For optimal western blotting of PLPPR1:

  • Sample preparation:

    • Prepare cell lysates in 2× SDS cell lysis buffer

    • Clarify lysates by centrifugation

    • Determine protein concentration using either the BCA method or Ionic Detergent Compatibility Reagent for Pierce 660nm Protein Assay

  • Gel electrophoresis conditions:

    • Load equal amounts of protein (20 μg/lane)

    • Use 10-12% SDS-PAGE gels for optimal resolution of PLPPR1 (~47 kDa)

  • Transfer parameters:

    • Transfer to PVDF membranes at 100V for 1 hour or 30V overnight

    • Verify transfer efficiency with Ponceau S staining

  • Blocking conditions:

    • Block membranes for 1 hour in 5% nonfat dried milk in either 0.1% Tween20 in PBS (PBST) or 0.1% Tween20 in 10 mM Tris–HCl pH 7.5 (TBST)

    • Alternatively, use 5% BSA in TBST for phospho-specific antibodies

  • Antibody incubation:

    • Incubate with primary antibodies overnight at 4°C

    • Wash thoroughly in PBST or TBST

    • Incubate with appropriate secondary antibody for 30 minutes at room temperature

  • Detection recommendations:

    • Use sensitive chemiluminescent substrates such as KPL LumiGlo

    • For quantitative analysis, use digital imaging systems that provide linear dynamic range

What assays are recommended for studying PLPPR1's effects on RhoA and Rac1 activation?

To effectively study PLPPR1's effects on RhoA and Rac1 activation, the following assays are recommended:

  • RhoA and Rac1 pull-down assays:

    • Use Rhotekin-RBD beads (50 μg) for RhoA pull-down

    • Use PAK-PBD beads (10 μg) for Rac1 pull-down

    • Incubate 600 μg of protein with the appropriate beads at 4°C for 1 hour

    • Wash complexes and elute in 2× SDS sample buffer before SDS-PAGE

  • Experimental conditions:

    • Serum-starve cells for 16-18 hours before treatments

    • Treat cells with either FAFBSA (control), 10% serum, or LPA (16 μM) to stimulate RhoA/Rac1 activation

    • Process samples quickly after treatment (within 2 minutes for LPA treatment)

  • Controls for pull-down assays:

    • Positive control: Cells treated with GTPγS to load all GTPases with GTP

    • Negative control: Cells treated with GDP to maintain GTPases in inactive state

    • Expression control: Verify equal expression levels of EGFP or EGFP-PLPPR1 between treatment conditions

  • Alternative approaches:

    • FRET-based biosensors for real-time visualization of RhoA/Rac1 activity

    • Immunoblotting for phosphorylated downstream targets of ROCK (MLC, ERM, MYPT1)

    • Co-immunoprecipitation to assess RhoGDI interactions with RhoA/Rac1

  • Data analysis:

    • Normalize active GTPase levels to total GTPase expression

    • Analyze using one-way ANOVA with Tukey's post-hoc multiple comparison test

    • Consider reporting fold-change relative to control conditions

How can I distinguish between direct effects of PLPPR1 and secondary effects through its binding partners?

Distinguishing between direct and indirect effects of PLPPR1 requires carefully designed experiments:

  • Structure-function analysis:

    • Generate PLPPR1 mutants that selectively disrupt specific protein interactions

    • The C-terminal truncation mutant (PLPPR1ΔC43) can be used to determine if phenotypes are dependent on the C-terminus

    • Create point mutations in putative interaction domains

  • Temporal analysis:

    • Use inducible expression systems to monitor immediate versus delayed effects

    • Acute inhibition versus chronic knockdown approaches

  • Domain-specific interference:

    • Design competing peptides that mimic PLPPR1 interaction domains

    • Use cell-permeable peptides to disrupt specific protein-protein interactions

  • Binding partner manipulation:

    • Simultaneously knockdown or overexpress RhoGDI to determine if PLPPR1 effects are dependent on this interaction

    • Use RhoGDI mutants that cannot bind PLPPR1 but maintain other functions

  • In vitro reconstitution:

    • Purify components and determine direct biochemical interactions

    • Assess whether PLPPR1 directly alters RhoGDI binding to RhoA/Rac1 in a purified system

  • Pathway analysis:

    • Determine if PLPPR1 effects on RhoA/Rac1 activation are compromised when downstream components are inhibited

    • Test if PLPPR1 effects persist when upstream activators are bypassed

What are the most reliable quantification methods for cell morphology changes induced by PLPPR1?

For reliable quantification of PLPPR1-induced morphological changes:

  • Membrane protrusion analysis:

    • Use time-lapse imaging to capture dynamic changes

    • Quantify protrusion rate, persistence, and retraction frequency

    • Measure protrusion length and area using image analysis software

  • Cell shape parameters:

    • Determine cell spreading area, circularity, and aspect ratio

    • Quantify the number and length of filopodia/lamellipodia

    • Analyze cell polarity and leading edge dynamics

  • Cytoskeletal organization:

    • Perform immunostaining for F-actin, microtubules, and focal adhesions

    • Quantify stress fiber density, orientation, and distribution

    • Measure focal adhesion size, number, and turnover rate

  • Standardized reporting:

Morphological ParameterControl (EGFP)PLPPR1-expressingStatistical Analysis
Cell area (μm²)[value ± SEM][value ± SEM][p-value, test used]
Circularity index[value ± SEM][value ± SEM][p-value, test used]
Protrusion length (μm)[value ± SEM][value ± SEM][p-value, test used]
Focal adhesion density[value ± SEM][value ± SEM][p-value, test used]
  • Automated analysis workflows:

    • Use CellProfiler or similar software for unbiased high-throughput analysis

    • Implement machine learning approaches for complex morphological classification

    • Apply consistent thresholding and segmentation parameters across all samples

What controls are necessary when studying PLPPR1's role in overcoming inhibitory factors for neurite outgrowth?

When studying PLPPR1's ability to overcome inhibitory factors for neurite outgrowth, implement these essential controls:

  • Substrate controls:

    • Poly-L-lysine (PLL) only as permissive control

    • PLL + CSPGs as inhibitory substrate

    • Concentration gradient of inhibitory factors to establish dose-response relationship

  • Expression controls:

    • EGFP-only expressing neurons as baseline control

    • Multiple PLPPR1 expression levels to assess dose-dependent effects

    • Include PLPPR1 mutants (e.g., PLPPR1ΔC43) to identify domain-specific requirements

  • Pathway validation controls:

    • RhoA/ROCK pathway inhibitor (Y-27632) as positive control for overcoming inhibition

    • Constitutively active RhoA to determine if PLPPR1 effects can be overcome

    • RhoGDI knockdown to verify the involvement of this interaction partner

  • Temporal controls:

    • Fixed timepoints (24, 48, 72 hours) to distinguish initial growth from long-term effects

    • Time-lapse imaging to capture dynamic neurite behaviors

  • Cell-type controls:

    • Primary neurons from different regions (hippocampal, cortical, DRG)

    • Comparison of developmental stages (embryonic vs. postnatal)

    • Assessment in both neuronal cell lines and primary neurons

  • Statistical considerations:

    • Minimum of three independent experiments

    • Appropriate statistical tests (t-tests for two conditions, ANOVA for multiple conditions)

    • Normality testing before applying parametric statistics (Kolmogorov–Smirnov tests)

What are common pitfalls when using PLPPR1 antibodies for immunofluorescence?

Common pitfalls and solutions when using PLPPR1 antibodies for immunofluorescence include:

  • High background signal:

    • Increase blocking time (2-3 hours at room temperature or overnight at 4°C)

    • Use alternative blocking agents (5% BSA, 10% normal serum, commercial blockers)

    • Include 0.1-0.3% Triton X-100 in blocking and antibody solutions for better penetration

    • Perform more extensive washing steps (5-6 washes of 10 minutes each)

  • Weak or absent signal:

    • Optimize fixation conditions (4% PFA for 15-20 minutes at room temperature)

    • Test different antigen retrieval methods (citrate buffer, EDTA buffer)

    • Increase antibody concentration or incubation time (overnight at 4°C)

    • Use signal amplification systems (tyramide signal amplification, secondary antibody kits)

  • Non-specific binding:

    • Pre-adsorb antibody with fixed, permeabilized cells lacking PLPPR1 expression

    • Include additional blocking steps with 5% milk or specific blocking peptides

    • Test antibodies from different suppliers or different clones

    • Validate specificity using PLPPR1 knockout or knockdown controls

  • Signal variability between experiments:

    • Standardize all steps of the protocol (fixation time, antibody dilutions)

    • Prepare fresh fixatives for each experiment

    • Process all experimental conditions in parallel

    • Include internal positive controls in each experiment

How can I address inconsistent results in RhoA/Rac1 activation assays when studying PLPPR1?

For addressing inconsistencies in RhoA/Rac1 activation assays when studying PLPPR1:

  • Sample preparation issues:

    • Ensure complete serum starvation (16-18 hours) before LPA stimulation

    • Maintain precise timing for LPA treatment (exactly 2 minutes)

    • Process samples rapidly to prevent GTP hydrolysis during preparation

    • Keep all samples on ice after lysis to minimize enzymatic activity

  • Technical considerations:

    • Verify freshness and activity of GTPase-binding beads before each experiment

    • Use flash-frozen aliquots of LPA stored at -80°C to prevent degradation

    • Maintain consistent protein amounts (600 μg) for pull-down assays

    • Include loading controls for total RhoA/Rac1 in all immunoblots

  • Expression variables:

    • Confirm consistent PLPPR1 expression levels between experiments

    • Sort cells for equivalent GFP expression when using EGFP-PLPPR1

    • Consider the timing post-transfection (48-72 hours optimal)

    • Verify that control cells truly lack PLPPR1 expression using RT-PCR

  • Assay optimization:

    • Include positive controls (serum stimulation) in each experiment

    • Run parallel assays for both RhoA and Rac1 from the same lysates

    • Test multiple LPA concentrations (4, 8, and 16 μM) to establish dose-response

    • Consider complementary approaches (FRET biosensors, immunostaining for downstream targets)

What are promising research areas for understanding PLPPR1's role in neural regeneration?

Promising research directions for understanding PLPPR1's role in neural regeneration include:

  • In vivo models of neural injury:

    • Further investigation of PLPPR1's role in promoting axon regeneration after spinal cord injury

    • Testing PLPPR1 overexpression in models of traumatic brain injury and stroke

    • Development of conditional PLPPR1 knockout mice to study its necessity in regeneration

  • Molecular interaction network:

    • Comprehensive proteomics to identify the complete PLPPR1 interactome beyond RhoGDI

    • Investigation of potential cross-talk between PLPPR1 and other regeneration-associated genes

    • Exploration of PLPPR1's interactions with extracellular matrix proteins and receptors

  • Therapeutic applications:

    • Development of small molecule mimetics that recapitulate PLPPR1's effects on RhoA/Rac1 signaling

    • Testing viral vector-mediated PLPPR1 overexpression for therapeutic purposes

    • Combination approaches with other regeneration-promoting strategies

  • Translational studies:

    • Examination of PLPPR1 expression and function in human neural tissues

    • Correlation of PLPPR1 genetic variants with recovery outcomes after neural injury

    • Investigation of PLPPR1's role in neurodegenerative diseases

How might PLPPR1 interact with other signaling pathways beyond RhoA/Rac1?

Potential interactions of PLPPR1 with other signaling pathways beyond RhoA/Rac1:

  • Other small GTPases:

    • Investigate potential effects on Cdc42, which often works in coordination with RhoA and Rac1

    • Examine possible interaction with Rap1, which plays roles in cell adhesion and polarization

    • Study effects on RhoB and RhoC, which have distinct functions from RhoA

  • Receptor tyrosine kinase signaling:

    • Explore potential cross-talk with growth factor receptors important in neural development

    • Investigate whether PLPPR1 affects MAPK pathway activation downstream of growth factors

    • Study potential interactions with neurotrophin receptor signaling

  • Cytoskeletal regulatory pathways:

    • Examine effects on cofilin phosphorylation status and activity

    • Investigate interactions with formins and the Arp2/3 complex

    • Study potential influence on microtubule dynamics and stability

  • Phospholipid signaling:

    • Given PLPPR1's relationship to lipid phosphatases, explore effects on PIP2/PIP3 balance

    • Investigate potential modulation of phospholipase C and phospholipase D signaling

    • Examine cross-talk with sphingolipid signaling pathways

  • Integration with calcium signaling:

    • Study whether PLPPR1 affects calcium-dependent processes in neurons

    • Investigate potential interactions with calcium-dependent kinases or phosphatases

    • Examine effects on calcium channel distribution or activity

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.