PTEN Human, His is a recombinant form of the human phosphatase and tensin homolog (PTEN) protein fused with a polyhistidine (His) tag at its N-terminus. This engineered variant facilitates purification and functional studies of PTEN, a critical tumor suppressor involved in regulating cell proliferation, survival, and genomic stability .
PTEN Human, His is produced in E. coli systems, ensuring cost-effective scalability. Key steps include:
Cloning: The PTEN coding sequence (residues 1–403) is fused with a His tag and inserted into an expression vector .
Expression: Induced under optimized conditions to maximize soluble protein yield .
Purification: The His tag binds to nickel columns, enabling high-purity isolation .
Formulation: Stabilized in Tris-HCl buffer (pH 8.0) with 20% glycerol, 1 mM EDTA, and 2 mM DTT to maintain enzymatic activity .
Substrate Specificity: Preferentially dephosphorylates PIP3, reducing Akt activation and suppressing oncogenic signaling .
Catalytic Activity: Retains dual phosphatase functionality (lipid and protein substrates) critical for tumor suppression .
Thermal Stability: Maintains activity in storage conditions (-80°C) for >12 months .
Enzymatic Activity | Assay Results |
---|---|
PIP3 Dephosphorylation | Specific activity: 0.5–1.0 µmol/min/mg |
Optimal pH | 7.5–8.5 |
Inhibitors | Vanadate, bpV(phen) (competitive inhibitors) |
PTEN Human, His is widely used in:
Cancer Biology: Studying PTEN’s role in tumor suppression and its interaction with PI3K/Akt/mTOR pathways .
Drug Discovery: Screening for compounds that modulate PTEN activity or stability .
Structural Studies: Investigating PTEN’s membrane-binding mechanisms via X-ray crystallography and NMR .
Key Findings Using PTEN Human, His:
Hemizygous PTEN loss in tumors leads to reduced protein levels (≤50%), accelerating cancer progression by dysregulating immune and metabolic pathways .
PTEN-Long, a secreted isoform with an extended N-terminus, exhibits tumor-suppressive effects in vivo by antagonizing Akt signaling .
PTEN functions primarily as a lipid and protein phosphatase that negatively regulates the PI3K/AKT/mTOR signaling pathway. The PTEN enzyme attaches (binds) to another PTEN enzyme (dimerizes) then binds to the cell membrane where it removes phosphate groups from phosphatidylinositol-3,4,5-trisphosphate (PIP3), converting it to phosphatidylinositol-4,5-bisphosphate (PIP2) . This dephosphorylation prevents activation of AKT, thus inhibiting downstream signaling that would otherwise promote cell proliferation and survival.
PTEN's tumor suppressive functions include:
Regulation of cell cycle progression
Induction of apoptosis through multiple pathways
Maintenance of genomic stability
Control of cell migration and tissue invasion
Inhibition of angiogenesis
The enzyme is part of a chemical pathway that signals cells to stop dividing and can trigger programmed cell death (apoptosis) . Additionally, it helps control cell movement, adhesion to surrounding tissues, and the formation of new blood vessels, all contributing to its role in preventing uncontrolled cell proliferation that leads to tumors .
PTEN comprises several functional domains that work together to enable its tumor suppressor activities:
N-terminal phosphatase domain: Contains the catalytic site responsible for dephosphorylating PIP3
C2 domain: Mediates binding to phospholipid membranes
C-terminal tail: Contains multiple phosphorylation sites that regulate PTEN stability and activity
PDZ-binding domain: Mediates protein-protein interactions with PDZ domain-containing proteins
For proper function, PTEN must dimerize and bind to cell membranes where it can access its lipid substrates . This dimerization is critical for PTEN's enzymatic activity, as highlighted in recent research. Mutations affecting any of these domains can impair PTEN function and contribute to disease development.
PTEN is a central negative regulator of the PI3K/AKT/mTOR pathway. When functioning normally:
Growth factors activate PI3K, which phosphorylates PIP2 to produce PIP3
PTEN counteracts this by dephosphorylating PIP3 back to PIP2
In the absence of PIP3, AKT remains inactive
Inactive AKT cannot phosphorylate downstream targets including TSC1/2 and PRAS40
Without these phosphorylation events, mTOR complex 1 (mTORC1) activity is restrained
When PTEN is inactive or absent, PIP3 accumulates, leading to hyperactivation of AKT and downstream effectors that promote cell proliferation, metabolism, survival, and growth . This dysregulation is a hallmark of many cancers and developmental disorders. The finely tuned balance between PI3K and PTEN activities is critical for maintaining normal cellular homeostasis.
PTEN Hamartoma Tumor Syndrome (PHTS) is a genetic condition caused by germline mutations in the PTEN gene. It encompasses several previously described clinical syndromes including Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome . PHTS is characterized by the development of multiple hamartomas (benign tissue overgrowths) throughout the body and a significantly increased risk for certain cancers .
Common features of PHTS include:
Non-cancerous hamartomas in various tissues
Macrocephaly (larger-than-average head size)
Skin abnormalities including trichilemmomas and oral papillomas
Developmental delay or autism spectrum disorder in some patients
The lifetime cancer risks for individuals with PHTS are substantial:
Type of Cancer | Lifetime Risk |
---|---|
Breast (in women) | 85% |
Thyroid | 35% |
Renal cell (kidney) | 35% |
Endometrium (uterus) | 28% |
Colorectal | 9% |
Melanoma | >5% |
These cancers generally occur in adults with PHTS, with the average age of diagnosis being approximately 30-50 years old . Thyroid cancer is an exception, as it sometimes occurs in children with the syndrome.
PTEN plays crucial roles in both innate and adaptive immunity. Patients with germline PTEN mutations exhibit a spectrum of immune dysfunction, ranging from asymptomatic lymphopenia to lymphoid hyperplasia, autoimmunity, and immunodeficiency .
Key immunological effects of PTEN mutations include:
T cell abnormalities:
Lower activation thresholds and hyperproliferation
Enhanced cytokine production
Altered development with expanded memory T cell populations
Impaired peripheral tolerance
B cell abnormalities:
Hyperactive B cell receptor signaling
Increased antibody production
Expanded germinal center responses
Altered transitional B cell frequencies
Clinical manifestations:
Thymic hyperplasia
Lymphadenopathy
Autoimmunity in some patients
Increased susceptibility to infections in others
In PHTS, PTEN expression is approximately 60% of normal in activated T cells, resulting in modest increases in PI3K signaling . This creates a threshold effect where the degree of PI3K pathway activation correlates with the severity of immune dysfunction.
PTEN mutations have been increasingly recognized as genetic risk factors for autism spectrum disorders (ASDs), particularly those accompanied by macrocephaly. While not explicitly detailed in the search results, scientific literature indicates that approximately 10-20% of individuals with ASDs and macrocephaly harbor germline PTEN mutations.
The mechanisms linking PTEN dysfunction to neurodevelopmental disorders include:
Dysregulated neuronal growth and proliferation
Abnormal synaptic formation and plasticity
Altered neuronal migration and circuit development
Imbalances in excitatory/inhibitory signaling
Mouse models with neuron-specific Pten deletion demonstrate features reminiscent of ASDs, including:
Social interaction deficits
Repetitive behaviors
Macrocephaly with enlarged neurons
Dendritic overgrowth and abnormal connectivity
These findings highlight the critical role of PTEN-mediated PI3K pathway regulation in normal neurodevelopment and suggest potential therapeutic targets for ASDs associated with PTEN mutations.
Comprehensive detection and characterization of PTEN mutations require multiple complementary technologies:
DNA-based methods:
Next-generation sequencing (NGS) for point mutations and small insertions/deletions
Multiplex Ligation-dependent Probe Amplification (MLPA) for detecting large genomic rearrangements and exon-level deletions
Sanger sequencing for confirmation of variants
Methylation analysis for epigenetic silencing
Protein-based methods:
Immunohistochemistry to evaluate PTEN protein expression in tissues
Western blotting for quantitative assessment of PTEN protein levels
Phosphatase activity assays to assess functional consequences of mutations
For clinical diagnostic purposes, a tiered approach is recommended:
First tier: NGS panel with PTEN coding region coverage plus MLPA
Second tier: Promoter region analysis
Third tier: Functional assessment through protein-based methods
Approximately 10% of PTEN mutations are large deletions that would be missed by sequencing alone , emphasizing the importance of comprehensive testing strategies that can detect multiple types of genetic alterations.
Expression and purification of active His-tagged PTEN protein involves several critical considerations:
Expression system selection:
Bacterial expression (E. coli BL21(DE3) or Rosetta strains) using pET vector systems
Eukaryotic expression (insect or mammalian cells) for studies requiring post-translational modifications
Construct design:
Full-length PTEN (403 amino acids) or specific domains
N-terminal or C-terminal 6xHis tag (N-terminal often preferred to avoid interference with C-terminal functional domains)
Inclusion of a protease cleavage site (TEV or PreScission) for tag removal if needed
Codon optimization for the expression host
Expression conditions:
Induction at lower temperatures (16-18°C) to improve solubility
Reduced IPTG concentration (0.1-0.5 mM) for slower expression
Co-expression with chaperones to improve folding
Purification strategy:
Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin
Ion exchange chromatography (typically Q-sepharose)
Size exclusion chromatography for final polishing
Buffer optimization: 25-50 mM Tris or HEPES (pH 7.4-8.0), 100-300 mM NaCl, reducing agents (DTT or TCEP), and 5-10% glycerol for stability
Quality control:
SDS-PAGE and western blotting for purity assessment
Phosphatase activity assays using artificial substrates (pNPP) or PIP3
Thermal shift assays to evaluate protein stability
PTEN has a tendency to aggregate, particularly at high concentrations, so careful attention to buffer conditions and storage is essential for maintaining enzymatic activity.
Multiple model systems have been developed for investigating PTEN function in different contexts:
Cellular models:
Isogenic cell line pairs with and without PTEN
CRISPR/Cas9-engineered PTEN knockout or knock-in cell lines
Patient-derived cells (lymphocytes, fibroblasts, or iPSCs)
3D organoid models that better recapitulate tissue architecture
Mouse models:
Germline Pten heterozygous mice that model PHTS
Conditional tissue-specific knockout using Cre-loxP system:
T cell-specific models for studying immune dysfunction
Brain-specific models for neurological phenotypes
Tissue-specific models for various cancer types
Knock-in models with specific mutations found in patients
Other model organisms:
Zebrafish models for developmental studies and drug screening
Drosophila models for genetic interaction studies
Patient-derived xenografts for preclinical drug testing
Each model system has strengths and limitations. For studying immune function, mouse models with conditional Pten deletion in specific immune cell populations have been particularly informative . For cancer studies, both genetically engineered mouse models and patient-derived models provide complementary insights.
The choice of model should be guided by the specific research question, with consideration of species-specific differences in PTEN regulation and function.
Targeting the PI3K pathway represents a genetically informed approach for treating conditions caused by PTEN deficiency. Several strategies have been developed:
Direct inhibition of pathway components:
PI3K inhibitors: Pan-PI3K inhibitors or isoform-specific inhibitors (particularly PI3Kδ inhibitors)
AKT inhibitors: Prevent phosphorylation and activation of downstream targets
mTOR inhibitors: Rapamycin (sirolimus) and rapalogs (everolimus, temsirolimus)
Dual PI3K/mTOR inhibitors: Target multiple nodes in the pathway
Implementation considerations:
Cell-type specificity: Different tissues show varying dependencies on PI3K signaling
Dosing strategies: Intermittent high-dose vs. continuous low-dose approaches
Combination approaches: Targeting multiple nodes or complementary pathways
Biomarkers for response: Phosphorylation status of pathway components
In PHTS patients, targeting mTOR via rapamycin has shown promise for normalizing thymic hyperplasia and correcting the frequency of transitional B cells . For immunological manifestations, PI3Kδ inhibitors have been evaluated as proof of concept in activated PI3K syndrome (APDS) and for use in polygenic immunological diseases .
Developing effective combination therapies for PTEN-deficient cancers requires understanding pathway interactions and resistance mechanisms:
Compensatory pathway targeting:
PTEN loss often activates compensatory signaling through the MAPK pathway
Combined inhibition of PI3K and MAPK pathways shows synergistic effects
Co-targeting cell cycle regulators (CDK4/6) can enhance efficacy
Synthetic lethality approaches:
PTEN loss creates dependencies on specific cellular processes
Identifying genes that are essential in PTEN-null but not PTEN-wild-type contexts
Examples include PARP inhibitors based on PTEN's role in DNA repair
Immunotherapy combinations:
PTEN loss can create an immunosuppressive tumor microenvironment
Combining PI3K inhibitors with immune checkpoint blockade
Strategies to enhance T cell infiltration and function
Experimental approaches for identifying combinations:
High-throughput drug screening in isogenic cell lines
In vivo testing in genetically engineered mouse models
Patient-derived xenograft models to capture tumor heterogeneity
Systems biology approaches to model pathway interactions
Biomarker development:
Identifying predictive biomarkers of response
Monitoring for resistance mechanisms
Real-time assessment of pathway inhibition
Rational combinations should target not only the primary consequence of PTEN loss (PI3K pathway activation) but also the secondary adaptations that emerge during treatment and contribute to resistance.
Based on the elevated cancer risks in PHTS, comprehensive surveillance protocols have been developed. These recommendations require a coordinated multidisciplinary approach :
Breast cancer surveillance (85% lifetime risk in women):
Annual mammography and breast MRI
Regular clinical breast examinations
Consideration of risk-reducing mastectomy in high-risk individuals
Thyroid cancer surveillance (35% lifetime risk):
Annual thyroid ultrasound examination
Thyroid function tests
Fine-needle aspiration of suspicious nodules
Renal cancer surveillance (35% lifetime risk):
Periodic renal imaging (ultrasound or MRI)
Urinalysis to check for hematuria
Endometrial cancer surveillance (28% lifetime risk in women):
Annual endometrial sampling or transvaginal ultrasound
Prompt investigation of abnormal bleeding
Colorectal cancer surveillance (9% lifetime risk):
Colonoscopy starting at age 35-40
Repeat every 3-5 years depending on findings
Skin cancer surveillance (>5% lifetime risk of melanoma):
Annual dermatological examination
Education about sun protection and self-examination
These guidelines are based primarily on expert opinion rather than robust clinical trial data, emphasizing the need for prospective evaluation of surveillance effectiveness in the PHTS population . Implementation should begin when the diagnosis of PHTS is made and continue throughout life, with consideration of individual risk factors and patient preferences.
PTEN functions within a complex network of tumor suppressor pathways with multiple points of cross-regulation:
p53 pathway: PTEN and p53 positively regulate each other's expression and function. PTEN stabilizes p53 by inhibiting MDM2-mediated degradation, while p53 can upregulate PTEN transcription, enhancing cell cycle arrest and apoptosis in response to cellular stress.
Rb pathway: PTEN loss activates AKT, which phosphorylates and inactivates GSK3β, leading to stabilization of cyclin D1 and inactivation of the Rb tumor suppressor, promoting cell cycle progression.
MAPK pathway: PTEN can negatively regulate the RAS/RAF/MEK/ERK pathway through multiple mechanisms, including direct dephosphorylation of adapter proteins and inhibition of integrin signaling.
Wnt/β-catenin pathway: PTEN antagonizes Wnt signaling by promoting β-catenin degradation, with PTEN loss potentially cooperating with Wnt pathway activation to drive tumor development.
Understanding these pathway interactions is crucial for developing combination therapeutic strategies. For example, combined loss of PTEN and p53 results in more aggressive tumors than loss of either gene alone, demonstrating synergistic effects that could be exploited therapeutically.
Beyond Mendelian disorders like PHTS, there is emerging evidence that PTEN plays a role in polygenic immune-mediated diseases . Reduced PTEN expression has been observed in multiple autoimmune conditions:
Systemic lupus erythematosus (SLE): Reduced PTEN levels in hyperreactive B cells correlate with disease activity .
Immune thrombocytopenia (ITP): PTEN is reduced in B cells, leading to hyperreactivity and increased plasma cell differentiation, as well as in autoreactive CD4 Th2 cells .
Immune-mediated nephritis: Decreased PTEN-L expression is associated with increased tissue inflammation .
Airway inflammatory disorders (asthma, COPD): Reduction in PTEN is linked to airway hyperresponsiveness and inflammation .
The reduced expression of PTEN in these disorders appears functionally significant rather than a mere bystander effect. Interestingly, while hundreds of loci have been associated with autoimmune disorders through genome-wide studies, PTEN itself has not emerged as a strong candidate locus . This suggests that observed transcriptional changes may be caused by trans-regulating loci or result from indirect regulation via epigenetic mechanisms.
These findings highlight PTEN's broader role in immune regulation beyond Mendelian PHTS and point to the PI3K pathway as a potential therapeutic target in immune-mediated inflammatory diseases.
PTEN activity, localization, and stability are tightly regulated through various post-translational modifications:
Phosphorylation:
The C-terminal tail contains multiple phosphorylation sites (Ser380, Thr382, Thr383, Ser385)
Phosphorylation by CK2, GSK3β, and other kinases stabilizes PTEN but reduces its activity
Dephosphorylation increases membrane association and phosphatase activity
Creates a "closed" conformation that protects from degradation but limits substrate access
Ubiquitination:
NEDD4-1, WWP2, and XIAP E3 ligases mediate PTEN ubiquitination
Monoubiquitination promotes nuclear import
Polyubiquitination targets PTEN for proteasomal degradation
Deubiquitinating enzymes (USP7, USP13) counteract this process
Acetylation:
PCAF and p300/CBP acetylate PTEN at lysine residues
Modulates PTEN interaction with PDZ domain-containing proteins
Affects protein stability and membrane localization
SUMOylation:
SUMO1 modification enhances PTEN phosphatase activity
Protects PTEN from ubiquitination
Influences subcellular localization
Oxidation:
Formation of disulfide bond between Cys124 and Cys71 inactivates PTEN
Occurs under oxidative stress conditions
Reversible modification regulated by redox environment
These modifications create a complex regulatory network that fine-tunes PTEN function in response to cellular context and external stimuli. Understanding this regulation provides insights into how PTEN activity might be altered in disease states and identifies potential therapeutic intervention points.
Several innovative approaches aim to restore PTEN function or compensate for its loss:
Gene therapy strategies:
Viral vector-mediated PTEN gene delivery to deficient cells
CRISPR-based approaches to correct specific PTEN mutations
mRNA therapeutics for temporary PTEN restoration
Protein-based approaches:
Delivery of recombinant PTEN protein variants with enhanced cell penetration
PTEN-Long: A translational variant that can be secreted and taken up by neighboring cells
Nanoparticle-mediated PTEN protein delivery
Small molecule interventions:
Compounds that stabilize remaining PTEN protein
Molecules that enhance PTEN catalytic activity
Targeting PTEN negative regulators like WWP1 (indole-3-carbinol compounds)
Pharmacological chaperones to correct folding defects in mutant PTEN
RNA-based therapeutics:
Anti-miRNA approaches to counter microRNAs that downregulate PTEN
siRNA targeting negative regulators of PTEN expression
Splice-switching oligonucleotides for mutations affecting PTEN splicing
Indirect approaches:
Synthetic lethality strategies exploiting dependencies created by PTEN loss
Compensatory phosphatase activation to counterbalance PI3K activity
These approaches face challenges including delivery to target tissues, achieving sufficient duration of effect, and potential immune responses. Combination strategies may ultimately prove most effective for restoring pathway homeostasis in PTEN-deficient conditions.
PTEN influences cellular metabolism and aging through several mechanisms that extend beyond its canonical tumor suppressor function:
Metabolic regulation:
Negative regulation of insulin signaling and glucose uptake
Control of fatty acid synthesis and lipid metabolism
Modulation of mitochondrial function and oxidative phosphorylation
Regulation of autophagy, a critical process for cellular recycling
Aging processes:
Protection against cellular senescence
Maintenance of stem cell populations and regenerative capacity
Regulation of reactive oxygen species and oxidative stress
Modulation of inflammatory processes associated with aging
Interaction with longevity-associated pathways (FOXO, SIRT1)
Age-related diseases:
Beyond cancer, PTEN has been implicated in cardiovascular disease
Emerging roles in neurodegenerative disorders like Alzheimer's and Parkinson's
Potential involvement in metabolic syndrome and type 2 diabetes
These connections between PTEN, metabolism, and aging represent fertile ground for future research, with potential implications for developing interventions that target age-related diseases through PTEN-associated pathways.
PTEN exhibits notable tissue-specific functions that could inform more targeted therapeutic strategies:
Tissue-specific signaling contexts:
Different tissues show varying dependencies on PI3K isoforms
Distinct feedback mechanisms operate in different cell types
Tissue-specific binding partners modulate PTEN activity
Implications for therapeutic targeting:
Isoform-selective PI3K inhibitors may provide tissue-selective effects
Dosing strategies could exploit tissue-specific sensitivity thresholds
Cell type-specific delivery systems could enhance therapeutic index
Biomarker development:
Tissue-specific PTEN-regulated gene signatures for patient stratification
Identification of compensatory mechanisms active in particular tissues
Monitoring tissue-specific pathway activation as response indicators
Personalized medicine approaches:
Patient-derived organoids to test drug responses ex vivo
Adaptive trial designs accounting for tissue-specific biomarkers
Combinatorial approaches tailored to individual molecular profiles
Research methodologies:
Single-cell techniques to resolve tissue heterogeneity
Spatial transcriptomics to understand PTEN function in tissue context
Systems biology approaches to model tissue-specific pathway interactions
Understanding these tissue-specific functions would allow for more precise therapeutic interventions that maximize efficacy while minimizing off-target effects, advancing the goal of truly personalized medicine for patients with PTEN-related disorders.
PTEN contains a tensin-like domain and a catalytic domain similar to that of dual-specificity protein tyrosine phosphatases . Unlike most protein tyrosine phosphatases, PTEN preferentially dephosphorylates phosphoinositide substrates . This activity is crucial for negatively regulating the intracellular levels of phosphatidylinositol-3,4,5-trisphosphate (PIP3), thereby inhibiting the AKT/PKB signaling pathway . The inhibition of this pathway is essential for controlling cell proliferation and survival, making PTEN a vital tumor suppressor .
Recombinant PTEN is often produced in various expression systems to study its function and for potential therapeutic applications . The human recombinant PTEN with a His Tag is a form of PTEN that has been genetically engineered to include a polyhistidine tag at the N-terminus . This His Tag facilitates the purification of the protein using affinity chromatography techniques, making it easier to isolate and study .
Mutations in the PTEN gene are associated with a wide range of cancers, including glioblastoma, lung cancer, breast cancer, and prostate cancer . The loss of PTEN function leads to uncontrolled cell growth and proliferation due to the unchecked activation of the AKT/PKB signaling pathway . Therefore, PTEN is a critical target for cancer research and therapeutic development .
Recombinant PTEN with a His Tag is widely used in research to understand its role in various cellular processes and its implications in cancer . By studying the recombinant protein, researchers can gain insights into the mechanisms of PTEN function and develop potential therapeutic strategies to restore its activity in cancer cells .