PINX1 Human

PIN2-Interacting Protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Function

Functional Mechanisms:

  • Telomerase inhibition: Directly binds hTERT and telomerase RNA (hTR), preventing telomere elongation .

  • Cell cycle regulation: Overexpression induces G0/G1 arrest and apoptosis, while knockdown accelerates G1/S transition .

  • Subcellular localization: Nuclear PINX1 associates with telomeres; nucleolar PINX1 mediates hTERT/TRF1 trafficking .

Tumor Suppressor Activity

Cancer TypePINX1 ExpressionClinical ImplicationsKey Findings
BreastReduced in 60-70% of tumorsIncreased tumorigenicityPinX1+/- mice develop mammary carcinomas
CervicalRegulated by p53Chemoresistance predictorHigh PINX1 correlates with paclitaxel sensitivity
EsophagealReduced in 50% of tumorsRadiation resistanceHigh PINX1 stabilizes telomeres, reducing RT efficacy
BladderInversely correlates with tumor stagePoor prognosisPINX1 knockdown increases proliferation and metastasis
OvarianLost in 33.8% of carcinomasLymph node metastasisPINX1 absent in metastatic cases

Mechanistic Insights:

  • Genomic instability: PinX1+/- mice exhibit telomere elongation, chromosomal fusions, and spontaneous carcinomas (liver, lung, GI tract) .

  • Pathway modulation:

    • Inhibits NF-κB to suppress MMP-2/9, reducing metastasis .

    • Activates p53-dependent apoptosis in HPV-associated cancers .

Clinical and Therapeutic Relevance

Diagnostic Markers:

  • Prognostic value:

    • Low PINX1 predicts poor survival in bladder (HR = 2.1) and esophageal cancers (HR = 1.8) .

    • High PINX1 correlates with paclitaxel response in cervical SCC (OR = 3.2) .

Therapeutic Targets:

  • Telomerase-dependent cancers: TID domain overexpression reduces tumorigenicity by 80% in xenograft models .

  • Radiosensitization: PINX1 knockdown enhances radiation-induced mitotic catastrophe in esophageal SCC .

Research Frontiers

  • Gene therapy: Adenoviral PINX1 delivery suppresses metastasis in renal cell carcinoma (70% reduction in lung nodules) .

  • Combination therapies: Co-targeting PINX1 and HSP90 enhances telomerase inhibition in breast cancer models .

  • Biomarker validation: Multi-center studies ongoing for PINX1 as a pan-cancer prognostic marker (NCT02859857, NCT03088787) .

Product Specs

Introduction
PINX1, a ubiquitously expressed protein, resides in nucleoli and telomere speckles. It harbors a Telomerase Inhibiting Domain capable of interacting with MCRS1, TERT, and TERF1. PINX1 exhibits potent telomerase inhibitory activity and potential tumor suppressor properties. Nucleolin recruits PINX1 to chromosome periphery, forming a complex essential for accurate chromosome congression. Moreover, PINX1 modulates the nucleolar accumulation and telomeric association of TRF1. Implicated in gastric cancer development, PINX1 expression serves as a marker for this malignancy. Constitutive PINX1 expression contributes to telomere maintenance via telomerase and tumorigenicity in cancerous cells.
Description
Recombinant Human PINX1, produced in E.Coli, is a single, non-glycosylated polypeptide chain consisting of 348 amino acids (1-328 a.a). With a molecular weight of 39.1 kDa, PINX1 is fused to a 20 amino acid His Tag at the N-terminus and purified using proprietary chromatographic methods.
Physical Appearance
Sterile, colorless solution.
Formulation
The PINX1 protein is supplied in a buffer containing 20mM Tris-HCl (pH 8), 1mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Minimize repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
PINX1, LPTL, LPTS, MGC8850, FLJ20565, Pin2-interacting protein X1, TRF1-interacting protein 1, Liver-related putative tumor suppressor, Protein 67-11-3.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MSMLAERRRK QKWAVDPQNT AWSNDDSKFG QRMLEKMGWS KGKGLGAQEH GATDHIKVQV KNNHLGLGAT INNEDNWIAH QDDFNQLLAE LNTCHGQETT DSSDKKEKKS FSLEEKSKIS KNRVHYMKFT KGKDLSSRSK TDLDCIFGKR QSKKTPEGDA SPSTPEENET TTTSAFTIQE YFAKRMAALK NKPQVPVPGS DISETQVERK RGKKINKEAT GKDVESYLQP KAKRHTEGKP ERAEAQERVA KKKSAPAEEQ LRGPCWDQSS KASAQDAGDH VQPPEGRDFT LKPKKRRGKK KLQKPVEIAE DATLEETLVK KKKKKDSK.

Q&A

What is PINX1 and what is its primary function in human cells?

PINX1 is an intrinsic telomerase inhibitor and putative tumor suppressor gene located on human chromosome 8p23, a region that exhibits the most frequent heterozygosity in common human adult epithelial malignancies . At the molecular level, PINX1 functions primarily as a negative regulator of telomerase activity, binding directly to the catalytic subunit of telomerase to inhibit its activity . This inhibition leads to telomere shortening, which plays a crucial role in regulating cellular senescence and preventing unlimited cellular proliferation . Research has demonstrated that overexpression of PINX1 inhibits telomerase activity, shortens telomeres, and can induce cellular crisis, whereas depletion of endogenous PINX1 increases telomerase activity and results in telomere elongation .

How is PINX1 expression measured in research settings?

PINX1 expression is commonly assessed at both the mRNA and protein levels using several complementary techniques:

  • Real-time quantitative PCR (RT-qPCR): This is the primary method used to quantify PINX1 mRNA expression, as demonstrated in studies of acute leukemia cells where researchers employed RT-qPCR with fluorescence probe hybridization to measure PINX1 mRNA levels .

  • Immunohistochemistry (IHC): Used to detect PINX1 protein expression in tissue samples, providing spatial information about expression patterns across different cell types within a tissue .

  • Western blotting: Applied to quantify PINX1 protein levels in cell lysates.

  • The Human Protein Atlas database: Serves as a reference resource that contains information regarding PINX1 expression profiles in human tissues at both mRNA and protein levels across 44 normal tissue types .

For reliability, researchers typically combine multiple methods to validate expression data, particularly when investigating differential expression between normal and malignant tissues.

What is the relationship between PINX1 and telomerase activity?

PINX1 demonstrates a direct inhibitory relationship with telomerase activity through several mechanisms:

  • PINX1 directly binds to the telomerase catalytic subunit (hTERT), inhibiting its enzymatic activity .

  • This inhibition prevents telomere elongation, maintaining telomeres at optimal length .

  • When PINX1 is depleted or downregulated, telomerase activity increases, leading to telomere elongation .

  • Conversely, overexpression of PINX1 inhibits telomerase activity, resulting in telomere shortening and potential cellular crisis .

This relationship is critical in cancer research because telomerase activation is observed in approximately 85-90% of human cancers, suggesting that PINX1 downregulation may be one mechanism through which cancer cells achieve telomerase activation and consequent replicative immortality . Methodologically, researchers investigate this relationship by manipulating PINX1 expression (through knockdown or overexpression) and then measuring the resulting changes in telomerase activity using telomeric repeat amplification protocol (TRAP) assays.

What evidence supports PINX1's role as a tumor suppressor?

Multiple lines of evidence support PINX1's classification as a tumor suppressor:

These findings collectively establish PINX1 as a significant tumor suppressor that functions primarily through regulation of telomerase activity and maintenance of chromosome stability.

How does PINX1 expression correlate with clinical outcomes across different cancer types?

PINX1 expression demonstrates significant associations with clinical outcomes across multiple cancer types, though these relationships can vary by cancer type:

PINX1 maintains chromosome stability through several interconnected molecular mechanisms:

  • Telomerase regulation: PINX1 directly inhibits telomerase activity, preventing excessive telomere elongation that can lead to telomere dysfunction and subsequent chromosome instability .

  • Telomere length maintenance: PINX1 is critical for maintaining telomeres at their optimal length. When PINX1 is depleted, resulting telomere elongation can paradoxically lead to telomere uncapping and chromosome fusion events .

  • Mitotic regulation: Beyond its telomerase inhibitory function, PINX1 plays an important role in mitosis. Its depletion can disrupt normal mitotic progression, leading to mitotic errors and consequent chromosome abnormalities .

  • DNA damage response: PINX1 affects the sensitivity of cancer cells to radiation-induced DNA damage, suggesting involvement in DNA repair mechanisms that maintain chromosome integrity .

  • Checkpoint function: Studies indicate that PINX1 may participate in cell cycle checkpoint regulation, ensuring proper chromosome segregation during cell division.

These mechanisms create a complex network through which PINX1 maintains chromosomal stability. When PINX1 expression is reduced, as observed in many cancers, the resulting chromosome instability likely contributes to the acquisition of additional oncogenic mutations and cancer progression .

How does the heterogeneity of PINX1 expression across different tissues impact research approaches?

The tissue-specific heterogeneity of PINX1 expression presents several methodological challenges for researchers:

PINX1 expression varies significantly across normal tissue types as documented in resources like The Human Protein Atlas, which provides mRNA and protein expression data across 44 normal tissue types . In cancer settings, PINX1 expression levels vary according to cancer cell types, creating additional complexity . This heterogeneity necessitates tailored research approaches including:

  • Tissue-specific control selection: Researchers must carefully select appropriate normal tissue controls that match the tissue of origin for the cancer being studied.

  • Multi-omics integration: Combining RNA-seq, protein expression data, and epigenetic profiling is essential to understand the full regulatory landscape controlling PINX1 in different tissues.

  • Single-cell analysis: Given potential cellular heterogeneity within tissues, single-cell approaches may be necessary to fully characterize PINX1 expression patterns and functions.

  • Tissue microenvironment considerations: Research must account for how the tumor microenvironment might influence PINX1 expression in specific tissues.

  • Validation across multiple datasets: Given expression variability, findings should be validated across multiple independent cohorts representative of the specific tissue being studied.

These considerations are particularly important when designing functional studies, as the impact of PINX1 manipulation may differ significantly between tissue types based on baseline expression levels and tissue-specific interaction partners.

What is the relationship between PINX1 expression and telomere dynamics in leukemia?

Studies on acute leukemia have provided specific insights into PINX1's relationship with telomere dynamics in hematological malignancies:

Researchers have investigated PINX1 expression in both acute non-lymphocytic leukemia (ANLL) and acute lymphoblastic leukemia (ALL) . Using real-time quantitative PCR with fluorescence probe hybridization, they measured PINX1 and hTERT mRNA expression in leukemia cells and during the differentiation of acute promyelocytic leukemia cells (NB4 cells) induced by all-trans retinoic acid (ATRA) .

The findings revealed a complex relationship between PINX1 expression, telomerase activity, and leukemic cell differentiation. During NB4 cell differentiation induced by ATRA, changes in PINX1 expression correlate with alterations in telomerase activity, suggesting that PINX1 may contribute to the regulation of telomerase during leukemic cell differentiation .

What are the most effective experimental approaches to manipulate PINX1 expression in cellular models?

Researchers studying PINX1 function employ several complementary approaches to manipulate its expression:

  • RNA interference (RNAi): Small interfering RNAs (siRNAs) or short hairpin RNAs (shRNAs) targeting PINX1 mRNA can be used to achieve transient or stable knockdown, respectively. This approach has been effectively used to demonstrate that depletion of endogenous PINX1 increases telomerase activity and elongates telomeres .

  • CRISPR-Cas9 gene editing: For complete knockout or precise mutation of PINX1, CRISPR-Cas9 provides advantages over RNAi by creating permanent genetic modifications. This approach is particularly valuable for studying long-term effects of PINX1 loss.

  • Overexpression systems: Plasmid-based overexpression of wild-type PINX1 or specifically its telomerase inhibitory fragment allows researchers to study gain-of-function effects. Studies have shown that overexpression inhibits telomerase activity, shortens telomeres, and can induce cellular crisis .

  • Inducible expression systems: Tetracycline-regulated or other inducible promoters enable temporal control of PINX1 expression, facilitating studies of acute versus chronic effects.

  • Domain-specific mutants: Structure-function studies can be conducted using constructs with mutations in specific PINX1 domains to dissect which regions are essential for different functions.

When designing these experiments, researchers should consider potential off-target effects, the efficiency of manipulation, and confirmation of altered expression at both mRNA and protein levels. Additionally, rescue experiments, where wild-type PINX1 is reintroduced following knockdown, provide important controls to confirm phenotype specificity.

How can researchers accurately determine the clinical significance of PINX1 expression patterns in patient samples?

Determining the clinical significance of PINX1 expression in patient samples requires rigorous methodological approaches:

  • Standardized tissue collection and processing: Consistent protocols for tissue acquisition, fixation, and storage are essential to minimize technical variability that could confound expression analysis.

  • Multi-modal expression analysis: Combining techniques such as immunohistochemistry, RT-qPCR, and in situ hybridization provides more comprehensive assessment than any single method alone.

  • Appropriate scoring systems: For immunohistochemical analysis, validated and reproducible scoring systems should be employed, considering both staining intensity and percentage of positive cells.

  • Proper statistical analysis: Statistical methods must account for confounding variables and multiple comparisons. Meta-analysis approaches have proven useful in establishing PINX1's prognostic value across cancer types (e.g., pooled hazard ratios for survival outcomes) .

  • Correlation with established markers: Analyzing PINX1 expression in relation to established clinical and molecular markers provides context for its significance.

  • Longitudinal sampling: When possible, analyzing samples across disease progression or treatment response provides insights into PINX1's dynamic role.

  • Large, well-characterized cohorts: Studies should utilize sufficiently powered cohorts with comprehensive clinical annotation to detect meaningful associations with outcomes.

What are the challenges in developing PINX1-based therapeutic approaches?

Developing therapeutic approaches based on PINX1 presents several significant challenges that researchers must address:

  • Delivery mechanisms: As a protein, PINX1 (or its telomerase inhibitory fragment) presents delivery challenges common to protein therapeutics, including cellular uptake, stability, and tissue-specific targeting.

  • Gene therapy considerations: For approaches aimed at restoring PINX1 expression in cancers with low expression, efficient gene delivery systems that target cancer cells specifically would be required.

  • Context-dependent effects: The variable expression and function of PINX1 across cancer types necessitates careful tailoring of therapeutic approaches to specific cancer contexts.

  • Integration with existing therapies: Researchers must determine how PINX1-based therapies would interact with standard treatments like chemotherapy, radiation, or targeted therapies.

  • Biomarker development: Identifying biomarkers that predict response to PINX1-based therapies would be essential for patient selection and monitoring.

  • Resistance mechanisms: Cancer cells may develop resistance to PINX1-based therapies through alternative pathways of telomerase activation or telomere maintenance.

  • Normal tissue toxicity: Since PINX1 functions in normal cells, potential impacts on healthy tissues must be carefully evaluated.

Despite these challenges, the research suggests potential therapeutic avenues, including the use of PINX1, especially its telomerase inhibitory fragment, as a means to inhibit telomerase in cancers where telomerase is activated . This approach could theoretically target a fundamental vulnerability in many cancer types while sparing normal cells with low telomerase activity.

How might single-cell analysis advance our understanding of PINX1's role in tumor heterogeneity?

Single-cell technologies offer transformative potential for understanding PINX1's role in tumor heterogeneity:

Current bulk tissue analysis methods can mask important cell-to-cell variations in PINX1 expression within tumors. Single-cell RNA sequencing (scRNA-seq) and CyTOF (cytometry by time-of-flight) approaches would enable researchers to:

  • Identify distinct subpopulations of cells with varying PINX1 expression levels within a tumor, potentially correlating these with other markers of stemness, proliferation, or treatment resistance.

  • Track changes in PINX1 expression during cancer evolution and in response to treatment at the single-cell level.

  • Discover cell type-specific PINX1 regulatory networks and downstream effects that might be obscured in bulk analyses.

  • Correlate PINX1 expression with telomere length at the single-cell level using techniques like telomere qFISH combined with PINX1 immunofluorescence.

  • Examine spatial relationships between cells with different PINX1 expression levels using spatial transcriptomics approaches.

These approaches could reveal whether PINX1 expression heterogeneity contributes to functional heterogeneity within tumors and identify cellular contexts where PINX1 loss is most consequential. Such insights could inform more precisely targeted therapeutic strategies and improve patient stratification for clinical trials of telomerase-targeting therapies.

What are the potential non-telomeric functions of PINX1 that warrant further investigation?

Emerging evidence suggests PINX1 may have important functions beyond telomerase inhibition:

  • Mitotic regulation: PINX1 has been implicated in mitosis, suggesting a broader role in cell cycle control that requires further characterization . Research should investigate how PINX1 interacts with the mitotic machinery and influences chromosome segregation.

  • DNA damage response: PINX1 affects cancer cell sensitivity to radiation-induced DNA damage . Further research should elucidate whether PINX1 directly participates in DNA repair pathways and how this function relates to its role in maintaining chromosome stability.

  • Signaling pathway interactions: Understanding how PINX1 intersects with established cancer-related signaling pathways could reveal new therapeutic targets or combination strategies.

  • Protein-protein interaction network: Comprehensive proteomics approaches are needed to map PINX1's interactome beyond telomerase components, potentially revealing novel functions.

  • Transcriptional regulation: Investigation into whether PINX1 influences gene expression patterns either directly or indirectly could unveil new regulatory roles.

  • Metabolic influences: Potential relationships between PINX1 and cellular metabolism have not been extensively explored but could be relevant to cancer's metabolic reprogramming.

These non-telomeric functions may explain why PINX1 appears to have context-dependent effects across different cancer types and could provide additional avenues for therapeutic intervention beyond telomerase inhibition.

How does the interplay between PINX1 and other telomere-associated proteins contribute to cancer development?

The complex interplay between PINX1 and other telomere-associated proteins represents a critical area for future research:

Telomere maintenance involves numerous proteins that work in concert, including components of the shelterin complex (TRF1, TRF2, POT1, TPP1, RAP1, and TIN2), telomerase components (TERT and TERC), and additional telomere-associated factors. PINX1 was originally identified as a TRF1-interacting protein (Pin2/TRF1 interacting protein) .

Future research should investigate:

  • How PINX1 cooperates with or antagonizes shelterin components to regulate telomere structure and function.

  • Whether alterations in multiple telomere-associated proteins, including PINX1, create synergistic effects on telomere dysfunction and chromosome instability.

  • The potential for compensatory mechanisms among telomere-associated proteins when PINX1 is dysregulated.

  • How post-translational modifications of PINX1 affect its interactions with telomerase and other telomere-associated proteins.

  • Whether cancer-associated mutations in telomere-related genes influence PINX1 function or expression.

This research will require advanced techniques including proximity labeling proteomics, super-resolution microscopy of telomeres, and systems biology approaches to model the complex interactions within the telomere interactome. Understanding these interactions could reveal vulnerable nodes for therapeutic targeting and explain variations in cancer susceptibility based on telomere biology.

Product Science Overview

Structure and Variants

PINX1 contains a telomerase inhibitory domain (TID), which is essential for its function. There are two known variants of PINX1, with the second variant lacking an exon in the 3’ coding region, resulting in a frameshift compared to variant 1. This frameshift leads to a shorter isoform with a distinct C-terminus .

Function and Mechanism

PINX1 interacts with several key proteins, including TERF1 (Telomeric Repeat-binding Factor 1), MCRS1 (Microspherule Protein 1), and TERT (Telomerase Reverse Transcriptase). By binding to these proteins, PINX1 can effectively inhibit telomerase activity, leading to telomere shortening and induction of cellular crisis. Over-expression of PINX1 results in decreased telomerase activity, while reduction of PINX1 leads to increased telomerase activity and elongation of telomeres .

Recombinant Human PINX1

Recombinant human PINX1 is produced using E. coli expression systems and is often tagged with a His-tag for purification purposes. The recombinant protein is typically used in research to study its function and interactions with other proteins. It is stored in a buffer containing Tris-HCl, DTT, and glycerol to maintain stability .

Applications in Research

Due to its role in telomere maintenance and potential as a tumor suppressor, PINX1 is a significant focus of cancer research. Studies involving recombinant human PINX1 help in understanding its mechanism of action and its potential therapeutic applications in cancer treatment .

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 2025 TheBiotek. All Rights Reserved.