PTP4A3 Human

Protein Tyrosine Phosphatase Type IVA Member 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Characteristics

PTP4A3 is a 173-amino acid protein with a catalytic phosphatase domain and a C-terminal prenylation motif that facilitates membrane localization . Key features include:

  • Enzymatic Activity: Dual-specificity phosphatase capable of dephosphorylating tyrosine, serine, and threonine residues .

  • Localization: Associates with plasma membranes and endosomes via prenylation, critical for its signaling functions .

  • Structural Homologs: Shares 97% sequence identity with murine and rat PRL-3, with conserved catalytic residues (e.g., Cys104, Arg201) .

Functional Roles in Cellular Signaling

PTP4A3 regulates diverse pathways involved in cell proliferation, migration, and survival:

PathwayMechanismBiological Impact
VEGF SignalingEnhances Src phosphorylation and endothelial cell migration .Promotes angiogenesis and tumor vascularization .
PI3K-AKT-ERKActivates AKT/ERK via PTEN regulation, driving cell cycle progression .Supports tumor growth and metastasis .
Rho GTPasesModulates cytoskeletal remodeling, facilitating epithelial-mesenchymal transition (EMT) .Increases invasiveness and metastasis .
NF-κB/JAK-STATUpregulates IL-1α and TGF-β1, influencing inflammation and immune evasion .Alters tumor microenvironment .

Oncological Relevance

PTP4A3 overexpression is observed in multiple cancers and correlates with poor prognosis:

Cancer-Specific Expression and Outcomes

Cancer TypeExpression LevelClinical CorrelationReferences
OvarianElevated in 79% of tumors; high in tumor endothelium .Poor survival; linked to angiogenesis .
RenalHigher in KIRC/KIRP; associated with poor DFS .Immune microenvironment dysregulation .
Uveal MelanomaDrives migration via CRMP2 dephosphorylation and MMP14 upregulation .Metastasis and reduced survival .
Breast/ColonAmplified in 33% of samples; reduces microvessel density in knockout models .Tumor-stromal interactions .

Preclinical Models

ModelFindingsImplications
Ptp4a3-null Mice30% reduced microvessel density in colon tumors; impaired VEGF-mediated permeability .Anti-angiogenic therapeutic potential .
RCC Cell LinesOverexpression increases proliferation, migration, and TGF-β1 expression .Targeting PTP4A3 may disrupt immune evasion .
T-ALLHigh expression promotes leukemia cell migration and survival .PTP4A3 inhibition as a therapeutic strategy .

Therapeutic Targets

  • Small-Molecule Inhibitors: Block PTP4A3 activity, reducing VEGF-driven endothelial migration .

  • Antibody-Based Therapies: PRL-3-zumab shows promise in disrupting tumor-stromal interactions .

Immune Microenvironment Modulation

PTP4A3 influences immune cell infiltration and cytokine production:

  • Immune Cell Correlations:

    • B Cells/CD8+ T Cells: High PTP4A3 expression associates with increased infiltration in renal/papillary cancers .

    • Treg Cells: Upregulates TGF-β1, enhancing immune suppression .

  • Cytokine Regulation: Induces IL-1α secretion via NF-κB/JAK-STAT pathways, promoting inflammation .

Product Specs

Introduction

Protein Tyrosine Phosphatase Type IVA Member 3 (PTP4A3), also known as PRL-3, is a prenylated protein tyrosine phosphatase involved in removing phosphate groups from tyrosine residues in proteins. It plays a crucial role in cellular processes like growth, movement, and invasion. PTP4A3 is known to accelerate the transition from the G1 to S phase during cell division. Primarily found in heart muscle cells and skeletal muscle, it is also present in the pancreas. Notably, PTP4A3 is frequently overexpressed in metastatic colon cancer and liver metastases originating from colorectal cancer. Its elevated expression is associated with tumor development and spread, making it a key player in the progression of colorectal, ovarian, liver, and skin cancers. PTP4A3 is believed to contribute to cardiac hypertrophy by hindering the mobilization of intracellular calcium in response to angiotensin II. Furthermore, its activity can be inhibited by compounds like sodium orthovanadate and peroxovanadium compounds.

Description
Recombinant human PTP4A3, expressed in E. coli, is a non-glycosylated polypeptide chain containing 193 amino acids (with an N-terminal 20 amino acid His tag). It has a molecular weight of 21.6 kDa. The protein is purified using proprietary chromatographic methods and is supplied in a non-glycosylated form.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PTP4A3 protein is supplied in a solution containing 20mM Tris-HCl buffer at pH 8.0, 2mM EDTA, 1mM DTT, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), the PTP4A3 solution can be stored at 4°C. For long-term storage, it is recommended to freeze the solution at -20°C. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is advisable. Repeated freezing and thawing should be avoided.
Purity
The purity of the PTP4A3 protein is determined by SDS-PAGE analysis and is greater than 95%.
Synonyms
Protein tyrosine phosphatase type IVA 3, Protein-tyrosine phosphatase 4a3, Protein-tyrosine phosphatase of regenerating liver 3, PRL-3, PRL-R, PTP4A3, PRL3.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MARMNRPAPV EVSYKHMRFL ITHNPTNATL STFIEDLKKY GATTVVRVCE VTYDKTPLEK DGITVVDWPF DDGAPPPGKV VEDWLSLVKA KFCEAPGSCV AVHCVAGLGR APVLVALALI ESGMKYEDAI QFIRQKRRGA INSKQLTYLE KYRPKQRLRFKDPHTHKTRC CVM.

Q&A

What is PTP4A3 and what is its significance in human cancers?

PTP4A3 is a member of the protein tyrosine phosphatase family located on chromosome 8q24.3, a region frequently amplified in various cancers. It promotes cellular invasion, motility, angiogenesis, and survival—properties associated with highly malignant and disseminated cancers . In high-grade serous ovarian cancer (HGSOC), PTP4A3 mRNA levels are 5–20 fold higher compared to nonmalignant cells .

Methodological approach to significance assessment:

  • Analyze bioinformatic databases (e.g., KMplot Survival Database) to correlate expression with patient outcomes

  • Compare PTP4A3 expression across cancer lineages using databases like the Cancer Cell Line Encyclopedia

  • Validate expression differences using qPCR and western blotting in appropriate cell line models

PTP4A3 overexpression significantly correlates with poor progression-free survival in epithelial ovarian cancer (p < 0.0001, hazard ratio = 1.35) . In early-stage HGSOC (stage I/II), patients with low PTP4A3 expression survived twice as long as those with high expression (81.2 vs. 44.3 months upper quartile survival) .

How is PTP4A3 expression regulated in human cells?

Unlike many oncogenes that undergo mutation, PTP4A3 overexpression typically results from:

  • Gene amplification - observed in 25% of HGSOC samples

  • Increased transcription and translation

  • Altered protein degradation pathways

  • Induction by genotoxic stress from chemotherapeutic agents like cisplatin, etoposide, and doxorubicin

  • Cytokine signaling - IL-6 induces PTP4A3 expression through STAT3 activation and SHP2 phosphatase repression

  • p53-mediated regulation - TP53 mutations (common in HGSOC) may impact PTP4A3 levels

Researchers should methodologically approach regulation studies by:

  • Performing copy number variation analysis to detect amplifications

  • Using reporter gene assays to identify transcriptional regulators

  • Employing pulse-chase experiments to measure protein stability

  • Assessing expression changes following cytokine or genotoxic stress exposure

What cellular processes does PTP4A3 regulate in cancer progression?

PTP4A3 coordinates multiple cellular processes contributing to malignancy:

  • Cell migration and invasion - PTP4A3 inhibition through JMS-053 causes concentration-dependent reduction in migration with EC50 of 250 nM in A2780 cells

  • Angiogenesis - PTP4A3 participates in VEGF signaling and contributes to pathological angiogenesis

  • Tumor cell survival - PTP4A3 promotes resistance to apoptosis

  • Drug resistance - Elevated expression is associated with chemoresistance

  • Cell adhesion alterations - PTP4A3 modifies cancer cell adhesion properties

Methodological approaches to study these processes:

  • Migration assays with quantitative endpoints (EC50 determination)

  • CRISPR/Cas9-mediated PTP4A3 depletion to validate functional roles

  • Pharmacological inhibition using selective compounds like JMS-053

  • In vivo tumor models comparing PTP4A3-expressing and PTP4A3-deficient conditions

How does PTP4A3 expression vary across different cancer types?

PTP4A3 shows distinct expression patterns across cancer lineages:

Cancer TypeRelative PTP4A3 ExpressionNotable Features
Leukemias/MyelomasHighest among 40 lineagesDetermined by RNAseq analysis
Ovarian Cancer13th highest lineage3.8-fold higher than average across human cancer lines
HGSOC subtypes5-20 fold higher than normalKuramochi cells show highest mRNA levels
Drug-resistant OvCaFurther elevatedCOV362-47R showed higher mRNA levels than parental COV362
Colon CancerSignificant expressionAssociated with reduced microvessel density in PTP4A3-null models

Methodological considerations for expression studies:

  • Normalize PTP4A3 expression to multiple housekeeping genes (HPRT, actin, GAPDH)

  • Compare expression across histological subtypes of the same cancer

  • Include drug-sensitive and drug-resistant paired cell lines

  • Validate mRNA findings with protein expression analysis

What methodologies are most effective for studying PTP4A3 function in cancer models?

Researchers should employ complementary approaches to comprehensively examine PTP4A3 function:

  • Genetic manipulation strategies:

    • CRISPR/Cas9-mediated depletion (achieved ~50% protein reduction in OVCAR4 cells)

    • Inducible knockdown systems for temporal control

    • Overexpression models mimicking gene amplification

  • Pharmacological inhibition:

    • JMS-053, a potent allosteric PTP4A3 inhibitor (EC50 ~250-500 nM for migration inhibition)

    • Structure-activity relationship studies using JMS-053 analogs

    • Target validation by demonstrating inhibitor ineffectiveness in PTP4A3-null cells

  • Functional assays:

    • Migration assays with and without IL-6 stimulation

    • 3D spheroid cytotoxicity assays for more physiologically relevant assessment

    • In vivo dissemination models to evaluate metastatic potential

  • Molecular analyses:

    • Quantitative proteomics to identify phosphorylation changes

    • Pathway analysis to understand signaling networks

    • Co-immunoprecipitation to identify protein interaction partners

How does PTP4A3 contribute to tumor microenvironment and angiogenesis?

PTP4A3's functions extend beyond cancer cells to influence the tumor microenvironment:

  • Endothelial cell effects:

    • PTP4A3 is expressed in tumor vasculature

    • Acts as a direct target of vascular endothelial growth factor (VEGF) signaling

    • Contributes to pathological angiogenesis as demonstrated by reduced microvessel density in Ptp4a3-null mouse models

  • Cytokine-mediated effects:

    • IL-6 markedly stimulates migration of OVCAR4 cells

    • CRISPR/Cas9-mediated PTP4A3 depletion reduces both basal and IL-6-stimulated migration

    • JMS-053 inhibits migration only in cells expressing PTP4A3, indicating target specificity

  • Methodological approaches:

    • Compare tumor vasculature in wild-type versus PTP4A3-null models

    • Analyze PTP4A3 expression in isolated tumor endothelial cells

    • Examine cytokine production and response in PTP4A3-manipulated models

    • Develop co-culture systems with tumor and stromal components

What are the challenges and strategies in developing effective PTP4A3 inhibitors?

Developing PTP4A3-targeted therapeutics presents several research challenges:

  • Current inhibitor landscape:

    • JMS-053 represents the most potent known allosteric small molecule PTP4A3 inhibitor

    • Structure-activity relationship studies of JMS-053 analogs have not yielded compounds with superior activity

    • JMS-053 showed greater potency than PARP inhibitors (olaparib, veliparib) in 3D cytotoxicity assays

  • Target validation approaches:

    • Demonstrate inhibitor activity is dependent on PTP4A3 expression

    • Compare phenotypic effects of pharmacological versus genetic PTP4A3 inhibition

    • Assess in vivo efficacy in reducing tumor dissemination

  • Combination strategies:

    • JMS-053 showed synergistic cytotoxicity with paclitaxel in OVCAR8 cells

    • Rational combination design based on complementary pathway targeting

    • Identification of synthetic lethal interactions with PTP4A3 inhibition

  • Translational considerations:

    • Biomarker development for patient selection

    • Pharmacokinetic optimization for in vivo activity

    • Toxicity assessment in normal versus malignant contexts

How can PTP4A3 expression patterns inform cancer prognosis and treatment strategies?

The prognostic and therapeutic implications of PTP4A3 expression provide valuable clinical insights:

  • Survival correlations:

    • High PTP4A3 expression associates with poor progression-free survival in epithelial ovarian cancer (p < 0.0001, HR = 1.35)

    • Early-stage HGSOC patients with low PTP4A3 expression survived twice as long as those with high expression

    • Advanced-stage patients (III/IV) also show significant survival differences based on PTP4A3 expression (p = 0.0027, HR = 1.29)

  • Genomic context analysis:

    • PTP4A3 highly expressed in 20% of HGSOC samples versus 13% for MYC in TCGA dataset

    • No coordinated co-expression between PTP4A3 and MYC despite proximity on chromosome 8q24

    • PTP4A3 amplification observed in 25% of HGSOC samples

  • Treatment implications:

    • PTP4A3 inhibition effective in chemoresistant ovarian cancer lines

    • Potential synergy with standard chemotherapeutics like paclitaxel

    • Patient stratification based on PTP4A3 expression might optimize therapeutic outcomes

What are optimal cell line models for studying PTP4A3 in ovarian cancer?

When selecting experimental models, researchers should consider:

Cell LineClassificationPTP4A3 ExpressionKey FeaturesAppropriate Applications
KuramochiHGSOCHighest mRNA levelsHighly representative of HGSOC Mechanistic studies, drug screening
OVCAR4HGSOCHigh protein levelsSuccessfully used for CRISPR/Cas9 PTP4A3 depletion Migration studies, IL-6 response
COV362HGSOCModeratePaired with resistant COV362-47R line Resistance mechanism studies
A2780/A2780cp20EndometrioidDetectableEstablished migration assay model Migration studies, EC50 determination
V581Patient-derived HGSOCNot specifiedClinically relevant primary model Translational studies
HIO-180Non-malignantLow (baseline)Appropriate control for expression studies Expression comparisons

Methodological considerations:

  • Match cell lines to specific research questions (e.g., chemoresistance, invasion)

  • Include appropriate controls (non-malignant, PTP4A3-low lines)

  • Validate key findings across multiple cell line models

  • Consider 3D culture systems for improved physiological relevance

How should researchers quantify and interpret PTP4A3-mediated effects on cell migration?

Migration assays are crucial for PTP4A3 functional studies:

What bioinformatic approaches best elucidate PTP4A3's clinical significance?

Researchers should employ multi-layered bioinformatic analyses:

  • Expression analysis workflow:

    • Compare PTP4A3 expression across cancer types using standardized datasets

    • Stratify patients by expression quartiles for survival analyses

    • Assess co-expression patterns with functionally related genes

  • Survival analysis methodology:

    • Generate Kaplan-Meier plots with appropriate statistical testing

    • Calculate hazard ratios with 95% confidence intervals

    • Perform multivariate analyses to control for confounding factors

  • Genomic integration approaches:

    • Analyze copy number alterations at the PTP4A3 locus (8q24.3)

    • Identify co-amplified genes that may functionally interact with PTP4A3

    • Correlate genomic events with transcriptomic and proteomic changes

  • Tool selection guidance:

    • KMplot Survival Database for prognostic assessment

    • TCGA data explorer for comprehensive genomic analysis

    • Cancer Cell Line Encyclopedia for model system selection

What are promising strategies for targeting PTP4A3 in combination therapies?

Based on current evidence, several combination approaches warrant investigation:

  • Chemotherapy combinations:

    • JMS-053 showed synergistic cytotoxicity with paclitaxel in OVCAR8 cells

    • Potential for overcoming resistance to standard chemotherapeutics

    • Rational scheduling to maximize therapeutic index

  • Targeted therapy pairings:

    • Combined inhibition of PTP4A3 and complementary oncogenic pathways

    • Exploration of synthetic lethal interactions with PTP4A3 overexpression

    • Targeting both tumor cells and supportive microenvironment components

  • Methodological approach to combination studies:

    • Systematic screening using drug combination matrices

    • Calculation of combination indices to quantify synergy

    • Validation in 3D models and in vivo systems

    • Investigation of mechanistic basis for observed synergies

  • Biomarker development for patient selection:

    • PTP4A3 expression or amplification status as primary biomarkers

    • Identification of additional markers that predict combination response

    • Development of clinically applicable testing methods

How might technological advances accelerate PTP4A3-targeted drug discovery?

Emerging technologies could significantly advance PTP4A3 inhibitor development:

  • Structure-based approaches:

    • Advanced computational modeling for allosteric inhibitor design

    • Fragment-based screening to identify novel chemical scaffolds

    • Protein-protein interaction targeting for indirect inhibition strategies

  • High-throughput phenotypic screening:

    • Migration-based high-content imaging assays

    • 3D spheroid penetration for assessing tissue distribution

    • Patient-derived organoid panels for clinically relevant screening

  • In vivo model innovations:

    • Genetically engineered mouse models with human-relevant PTP4A3 alterations

    • Patient-derived xenografts stratified by PTP4A3 expression

    • Metastatic models specifically addressing PTP4A3's role in dissemination

  • Translational acceleration strategies:

    • Repurposing existing drugs that modulate PTP4A3-dependent pathways

    • Development of companion diagnostics in parallel with therapeutics

    • Adaptive trial designs incorporating PTP4A3 biomarkers

What are key unresolved questions in PTP4A3 biology requiring further investigation?

Several fundamental questions remain to be addressed:

  • Biochemical mechanisms:

    • Identification and validation of direct physiological substrates

    • Characterization of PTP4A3's protein interaction network

    • Elucidation of structure-function relationships for phosphatase activity

  • Regulatory networks:

    • Comprehensive mapping of upstream regulators beyond IL-6 and p53

    • Understanding epigenetic control of PTP4A3 expression

    • Clarification of post-translational modifications affecting function

  • Therapeutic resistance mechanisms:

    • Adaptations to PTP4A3 inhibition in long-term treatment models

    • Compensatory pathway activation following PTP4A3 targeting

    • Patient-derived models of intrinsic and acquired resistance

  • Methodological approaches to address these questions:

    • Phosphoproteomic identification of substrate candidates

    • ChIP-seq analysis of transcription factor binding at the PTP4A3 locus

    • Long-term adaptation studies in cell line and patient-derived models

    • Systems biology approaches to model PTP4A3 network perturbations

Product Science Overview

Structure and Function

PTP4A3 is part of a small class of prenylated PTPs, which means it undergoes a post-translational modification where a lipid group is added to the protein. This modification is important for the protein’s localization to the cell membrane, where it can interact with other signaling molecules . The primary functions of PTP4A3 include:

  • Enhancing Cell Proliferation: PTP4A3 promotes the progression of cells from the G1 phase to the S phase of the cell cycle, thereby stimulating cell division .
  • Increasing Cell Motility: The protein enhances the ability of cells to move, which is crucial for processes such as wound healing and immune responses .
  • Promoting Invasive Activity: PTP4A3 is involved in the invasive behavior of cells, which is a characteristic feature of cancer metastasis .
Role in Cancer

PTP4A3 has been extensively studied for its role in cancer, particularly in metastasis. It has been found to be overexpressed in various types of cancers, including colorectal cancer, breast cancer, ovarian cancer, gastric cancer, esophageal squamous cell carcinoma, and hepatocellular carcinoma (HCC) . The overexpression of PTP4A3 is associated with:

  • Tumor Differentiation: Higher levels of PTP4A3 are correlated with poorly differentiated tumors, which tend to be more aggressive and have a worse prognosis .
  • Poor Prognosis: Patients with high PTP4A3 expression often have a poorer overall survival rate and a higher likelihood of cancer recurrence .
  • Metastasis: PTP4A3 plays a significant role in the metastatic spread of cancer cells to other parts of the body .
Clinical Implications

Given its role in cancer progression and metastasis, PTP4A3 is considered a potential prognostic marker and therapeutic target. High expression levels of PTP4A3 can serve as an indicator of poor prognosis in cancer patients, making it a valuable marker for predicting patient outcomes . Additionally, targeting PTP4A3 with specific inhibitors could potentially reduce cancer cell proliferation, motility, and invasiveness, offering a new avenue for cancer therapy.

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.