PTPN1 Human

Protein Tyrosine Phosphatase Non Receptor Type-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Structure and Function

PTPN1 is a 435-amino acid protein localized to the cytoplasmic face of the endoplasmic reticulum . Its catalytic domain contains three critical loops:

  • WPD loop (residue Asp181): Facilitates substrate binding and structural flexibility.

  • pTyr loop (residue Tyr46): Determines substrate selectivity by controlling access to the catalytic cleft.

  • Q loop (residue Gln262): Stabilizes intermediates during dephosphorylation .

The enzyme’s primary role involves dephosphorylating tyrosine residues on signaling molecules, including:

  • Insulin receptor (INSR)

  • Epidermal growth factor receptor (EGFR)

  • Janus kinase 2 (JAK2) .

Recent structural studies using room-temperature crystallography revealed allosteric sites that modulate PTPN1 activity, offering new targets for inhibitor design .

Mechanism of Action

PTPN1 operates via a two-step catalytic mechanism:

  1. Dephosphorylation: Cys215 nucleophilically attacks phosphotyrosine substrates, forming a thiophosphate intermediate.

  2. Hydrolysis: Gln262 and Asp181 position water to hydrolyze the intermediate, regenerating active enzyme .

Oxidative inactivation by reactive oxygen species (ROS) reversibly modifies Cys215 to sulfenic acid, providing a regulatory mechanism tied to cellular redox states .

Metabolic Disorders

  • Diabetes and Obesity: PTPN1 knockout mice exhibit enhanced insulin sensitivity and resistance to diet-induced obesity . By dephosphorylating INSR and leptin receptors, PTPN1 negatively regulates metabolic signaling, making it a therapeutic target for type 2 diabetes .

Cancer

PTPN1 exhibits dual roles in oncology:

Cancer TypeRole of PTPN1MechanismPrognostic Impact
Breast CancerOncogenicOverexpression with HER2; promotes proliferation and metastasis Shorter survival
GliomaOncogenicActivates MAPK/ERK and PI3K/AKT pathways Poor prognosis
LymphomaTumor-suppressiveLoss accelerates tumorigenesis in p53-deficient models Reduced survival

PTPN1 expression correlates with immune checkpoint markers (e.g., PD-L1) and modulates T cell activity, influencing immunotherapy responses .

Therapeutic Targeting

Inhibitor Development:

  • Allosteric inhibitors: Target distal sites linked to catalytic activity, reducing off-target effects .

  • Combination therapies: PTPN1 inhibition synergizes with anti-PD-1 treatments and CAR-T cell therapy .

Drug Sensitivity:

  • PTPN1 deletion enhances chemosensitivity to paclitaxel in breast cancer cells .

  • Small-molecule inhibitors (e.g., ABBV-CLS-484) show promise in preclinical trials .

Research Advancements

  • Immune Microenvironment: High PTPN1 expression associates with CD163+ M2 macrophage infiltration and reduced CD8+ T cell activity in breast cancer .

  • Structural Insights: Hydrogen-deuterium exchange studies identified dynamic regions essential for substrate recognition .

Product Specs

Introduction
Protein Tyrosine Phosphatase 1B (PTP1B) is a key regulator of cellular signaling pathways, playing a crucial role in processes like cell growth, differentiation, and response to stimuli such as insulin and interferon. As a member of the protein tyrosine phosphatase (PTP) family, it specifically removes phosphate groups from tyrosine residues on target proteins. PTP1B's involvement in insulin signaling is particularly noteworthy, as it dephosphorylates and negatively regulates the insulin receptor kinase, impacting glucose metabolism. Moreover, its interaction with other receptor tyrosine kinases like EGFR, and kinases like JAK2 and TYK2, highlights its multifaceted role in controlling cell growth and response to interferon.
Description
This product consists of the human recombinant Protein Tyrosine Phosphatase Non-Receptor Type 1, produced in E. coli. This single, non-glycosylated polypeptide chain comprises 321 amino acids, resulting in a molecular weight of 37.3 kDa.
Physical Appearance
A clear and colorless solution that has been sterilized by filtration.
Formulation
This protein solution has a concentration of 1 mg/ml and is formulated in a buffer containing 25mM Tris-HCl (pH 7.5), 2mM beta-mercaptoethanol, 1mM EDTA, 1mM DTT, and 20% glycerol.
Stability
For optimal storage, refrigerate at 4°C if the entire vial will be used within 2-4 weeks. For extended storage, freeze at -20°C. To ensure stability during long-term storage, consider adding a carrier protein like HSA or BSA (0.1%). Minimize repeated freezing and thawing to maintain product quality.
Purity
Determined by SDS-PAGE analysis, the purity is greater than 95%.
Biological Activity
The specific activity of this enzyme is measured as its ability to hydrolyze p-nitrophenyl phosphate (pNPP). Under standard assay conditions (pH 7.5, 37°C), it exhibits an activity greater than 10,000 units/mg, where one unit represents the hydrolysis of 1.0 nmole of pNPP per minute.
Synonyms
Tyrosine-protein phosphatase non-receptor type 1, EC 3.1.3.48, Protein-tyrosine phosphatase 1B, PTP-1B, PTPN1, PTP1B.
Source
Escherichia Coli.
Amino Acid Sequence
MEMEKEFEQI DKSGSWAAIY QDIRHEASDF PCRVAKLPKN KNRNRYRDVS PFDHSRIKLHQEDNDYINAS LIKMEEAQRS YILTQGPLPN TCGHFWEMVW EQKSRGVVML NRVMEKGSLKCAQYWPQKEE KEMIFEDTNL KLTLISEDIK SYYTVRQLEL ENLTTQETRE ILHFHYTTWPDFGVPESPAS FLNFLFKVRE SGSLSPEHGP VVVHCSAGIG RSGTFCLADT CLLLMDKRKDPSSVDIKKVL LEMRKFRMGL IQTADQLRFS YLAVIEGAKF IMGDSSVQDQ WKELSHEDLE PPPEHIPPPPRPPKRILEPHN.

Q&A

What is the molecular structure of PTPN1 and how does it influence substrate specificity?

The catalytic domain of PTPN1 features a deep cleft formed by three critical loops: the WPD loop containing Asp181, the pTyr loop with Tyr46, and the Q loop with Gln262. The pTyr loop and Tyr46 residue, located on the protein surface, determine substrate access depth, creating a structural basis for selectivity. Upon substrate binding, the WPD loop undergoes a conformational change, closing around the substrate and establishing stabilizing pi-stacking interactions between the phosphotyrosine substrate and Phe182 . This structure-function relationship is crucial for understanding how PTPN1 selectively dephosphorylates various substrates including insulin receptor, JAK2, and other signaling molecules.

How does PTPN1 regulate insulin signaling at the molecular level?

PTPN1 functions as a negative regulator of insulin signaling by dephosphorylating phosphotyrosine residues on the activated insulin receptor kinase . This dephosphorylation disrupts downstream signal transduction, dampening insulin's effects. Genetic studies in mice demonstrate that ablation of PTPN1 results in enhanced insulin sensitivity , providing direct evidence for its inhibitory role. Beyond insulin receptor, PTPN1 also dephosphorylates other components of the insulin signaling pathway, including IRS1 (Insulin Receptor Substrate 1), with which it has been shown to directly interact . This multilevel regulation positions PTPN1 as a master regulator of insulin responsiveness.

What is the epidemiological significance of PTPN1 variants in type 2 diabetes risk?

The PTPN1 gene increases type 2 diabetes risk by approximately 30%, and remarkably, about 35% of the population carries risk variants . Through genetic epidemiology calculations, this translates to an additional 3.6 million cases in total type 2 diabetes burden . The gene is located on the long arm of chromosome 20 (20q), a region consistently linked to diabetes inheritance across multiple genetic studies . Interestingly, the evidence suggests that disease association is mediated by DNA sequence differences outside the coding region of the PTPN1 gene rather than by coding variants, which are few and uncommon .

How do PTPN1 polymorphisms interact with environmental factors in obesity-related phenotypes?

Studies examining genetic-environmental interactions reveal that PTPN1 polymorphisms are significantly associated with adiposity markers in adolescents, but physical activity appears to modulate this genetic predisposition . This gene-environment interaction was identified through studies of 1057 European adolescents (aged 12-18) where adiposity markers, biochemical parameters, and objectively measured physical activity were assessed alongside PTPN1 genotyping . The findings suggest a mechanistic pathway through which physical activity positively influences obesity outcomes by potentially attenuating the effects of PTPN1 genetic variants, providing valuable insights for personalized intervention strategies.

What methodologies are most effective for investigating PTPN1's role in neuroblastoma?

In studying PTPN1's impact on neuroblastoma (NB), researchers employed a multi-method approach combining immunohistochemistry, gene knockdown, proliferation assays, phosphorylation analysis, and RT-qPCR on human NB cell lines (SH-SY5Y, SMS-KCNR, and IMR-32) and 44 human NB tumor samples . This comprehensive methodology revealed that PTPN1 knockdown in SH-SY5Y cells resulted in increased tyrosine phosphorylation and cell proliferation . Critically, high PTPN1 expression in NB tumors correlated with metastasis and poor prognosis , identifying it as both a potential regulator of NB cell growth and a prognostic biomarker, demonstrating the value of integrating multiple experimental approaches in cancer research.

What structural characterization techniques provide the most insight into PTPN1 variant functions?

The most comprehensive approach to characterizing PTPN1 variants combines room-temperature X-ray crystallography, hydrogen-deuterium exchange mass spectrometry, and computational modeling . This multi-technique strategy revealed how human variants modulate PTP1B's three-dimensional structure through distinct allosteric conduits that energetically link distal, highly ligandable structural regions to the active site . In one study, researchers selected 12 rare variants from exomes of 997 people with persistent thinness and 200,000 people from UK Biobank, finding that seven variants impaired PTP1B function, increasing leptin-stimulated STAT3 phosphorylation in cells . This approach provided critical insights for designing allosteric PTP1B inhibitors for potential obesity treatment.

What are the optimal experimental models for studying PTPN1 in hematopoietic disorders?

For investigating PTPN1 in hematopoietic contexts, conditional knockout models with bone marrow transplantation assays have proven most informative. Researchers generated Mx1-Cre;Ptpn1fl/fl mice and induced Ptpn1 deletion using polyinosine-polycytosine (pI-pC) injections . This approach demonstrated that Ptpn1 deletion increased white blood cell and neutrophil counts, expanded myeloid lineages, and eventually led to myelofibrosis . To distinguish cell-autonomous effects, cell-autonomous bone marrow transplantation assays were performed, wherein control and Ptpn1-knockout bone marrow was transplanted into irradiated recipients . Additionally, competitive repopulation assays revealed that Ptpn1 deletion increased hematopoietic stem cell reconstitution capacity and promoted myeloid lineage expansion . These models collectively provide robust systems for dissecting PTPN1's role in hematopoietic regulation.

How can researchers effectively address the oxidation sensitivity of PTPN1 in experimental settings?

The catalytic cysteine residue (Cys215) of PTPN1 is highly susceptible to oxidation, which can compromise experimental reliability and physiological relevance. Research shows that in cancer cell lines like HepG2 and A431, up to 40% of Cys215 residues can be irreversibly oxidized under cellular conditions . To address this challenge, researchers should:

  • Conduct experiments under controlled redox conditions with appropriate antioxidants

  • Implement rapid protein extraction protocols that minimize oxidation time

  • Use mutant forms of PTPN1 with oxidation-resistant catalytic sites for mechanistic studies

  • Employ activity-based probes that can distinguish between active and oxidized forms

  • Quantify the oxidation state using mass spectrometry before activity measurements

These methodological considerations are particularly critical when comparing PTPN1 activity across different disease states with varying redox environments.

What approaches can resolve contradictory findings regarding PTPN1's role in different cancer types?

The apparently contradictory roles of PTPN1 in cancer (tumor-promoting in breast cancer versus tumor-suppressive in hematopoietic malignancies) require careful experimental design to resolve. Effective approaches include:

  • Tissue-specific conditional knockout models to isolate effects in specific cellular contexts

  • Temporal control of PTPN1 modulation to distinguish roles in initiation versus progression

  • Substrate-specific mutants to identify which downstream pathways mediate different outcomes

  • Analysis of post-translational modifications that may alter PTPN1 function

  • Integration of genomic, proteomic, and phosphoproteomic data to map context-specific signaling networks

This systematic approach can help reconcile disparate findings by identifying the specific molecular contexts that determine whether PTPN1 acts as an oncogene or tumor suppressor in a given cancer type.

How might PTPN1 inhibition strategies be optimized for dual metabolic and immune benefits?

Recent identification of PTPN1 as both a metabolic regulator and immune checkpoint presents unique opportunities for therapeutic development. Optimization strategies should:

  • Develop tissue-selective inhibitors that can target specific cell populations (e.g., adipocytes versus T cells)

  • Investigate the dose-response relationship to identify therapeutic windows where immune activation occurs without compromising metabolic benefits

  • Explore combination therapies with established immune checkpoint inhibitors or metabolic agents

  • Design time-release formulations that can address the temporal aspects of both metabolic and immune pathways

  • Identify biomarkers that predict which patients might benefit most from dual-action PTPN1 targeting

This approach could be particularly valuable in metabolic-associated conditions with inflammatory components, such as non-alcoholic steatohepatitis or certain cancer types where both metabolism and immunity play crucial roles.

What potential does single-cell multiomics hold for advancing PTPN1 research?

Single-cell multiomics approaches offer unprecedented opportunities to understand PTPN1 biology across heterogeneous cell populations. Future research should leverage:

  • Integrated single-cell RNA/protein analysis to correlate PTPN1 expression with its activity state

  • Spatial transcriptomics to map PTPN1 function within complex tissue architectures

  • Single-cell phosphoproteomics to identify cell-type specific PTPN1 substrates

  • Temporal single-cell profiling to track PTPN1-mediated signaling dynamics during disease progression

  • Multi-parameter CyTOF analysis to correlate PTPN1 activity with immune cell phenotypes and exhaustion markers

Evidence from liver cancer studies already demonstrates the value of single-cell analysis, which revealed PTPN1 enrichment in specific T cell populations linked to exhaustion phenotypes , a finding impossible to detect in bulk tissue analysis.

Product Science Overview

Gene and Protein Structure

PTPN1 is encoded by the PTPN1 gene in humans . The gene is located on chromosome 20 and has been extensively studied due to its significant role in various cellular processes . The protein itself is composed of several domains, including a highly conserved catalytic domain that is essential for its enzymatic activity . The catalytic site of PTP1B is located within a deep cleft formed by three loops, which include the WPD loop with the Asp181 residue, a pTyr loop with the Tyr46 residue, and a Q loop with the Gln262 residue .

Biological Function

PTP1B is primarily localized to the cytoplasmic face of the endoplasmic reticulum . It plays a crucial role in regulating several signaling pathways by dephosphorylating key tyrosine-phosphorylated proteins. One of its most well-known functions is as a negative regulator of the insulin signaling pathway . By dephosphorylating the insulin receptor kinase, PTP1B modulates insulin sensitivity and glucose homeostasis . This makes it a promising therapeutic target for the treatment of type 2 diabetes and obesity .

Role in Disease

PTP1B has been implicated in various diseases, including type 2 diabetes, obesity, and cancer . In the context of cancer, PTP1B has been identified as an oncogene and a potential therapeutic target . It is involved in the dephosphorylation of several tyrosine kinases, such as the epidermal growth factor receptor (EGFR), insulin-like growth factor 1 receptor (IGF-1R), and Janus kinase 2 (JAK2), which are critical for cell growth and proliferation .

Therapeutic Potential

Given its role in regulating insulin signaling and its involvement in cancer, PTP1B is considered a valuable target for drug development . Inhibitors of PTP1B are being explored as potential treatments for type 2 diabetes, obesity, and certain types of cancer . Research is ongoing to develop selective and potent inhibitors that can effectively modulate PTP1B activity without causing adverse effects .

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.