PTTG1 Human

Pituitary Tumor-Transforming Protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Protein Characteristics

PTTG1 is a 222-amino-acid polypeptide (24.1 kDa) with distinct functional domains:

FeatureDescriptionSource
PXXP MotifsTwo proline-rich motifs critical for transforming/tumorigenic activities and growth factor stimulation (e.g., bFGF).
Destruction Box (D-box)Enables ubiquitination by the anaphase-promoting complex (APC) and subsequent proteasomal degradation.
Acidic C-Terminal DomainFunctions as a transactivation domain, facilitating transcriptional regulation.
Subcellular LocalizationPrimarily cytosolic, with partial nuclear localization and association with structures like the Golgi apparatus and microtubules.

Interactions

PTTG1 binds to proteins involved in mitosis, DNA repair, and apoptosis:

  • DNAJA1: Chaperone involved in protein folding.

  • Ku70: DNA repair protein.

  • p53: Negatively regulates p53-mediated apoptosis.

  • Separase (ESPL1): Inhibits premature sister chromatid separation during mitosis. |

Mitotic Regulation

PTTG1 acts as a securin homolog, binding to separase (ESPL1) to block its proteolytic activity on cohesin complexes. This prevents premature sister chromatid separation until anaphase onset, ensuring genomic stability. Its degradation via APC-mediated ubiquitination releases ESPL1, enabling chromosomal segregation. |

DNA Repair and Apoptosis

  • DNA Damage Response: Interacts with Ku70 to facilitate DNA repair.

  • p53 Regulation: Inhibits p53 transcriptional activity, reducing apoptosis and promoting survival of damaged cells. |

Oncogenic Pathways

PTTG1 drives tumorigenesis through:

PathwayMechanismCancer Type
Epithelial-Mesenchymal Transition (EMT)Activates MMP-2 expression, enhancing cell migration and invasion.Breast, Lung, Glioma
PI3K/AKT and MAPKPromotes proliferation and survival via kinase signaling.Melanoma, Prostate

Overexpression and Prognostic Impact

PTTG1 is overexpressed in >70% of cancers, including:

Cancer TypeExpression LevelClinical OutcomeSource
Multiple Myeloma (MM)36–70% overexpressionPoor survival (HR = 2.49, p = 0.0075).
GliomaElevated vs. normalCorrelates with tumor grade.
Breast CancerHigh expressionEnhanced metastasis and drug resistance.

Therapeutic Targeting

  • siRNA Knockdown: Reduces tumor growth in MM and breast cancer models. In vitro studies show decreased proliferation (e.g., MTT assay) and downregulation of oncogenes like CCNB1 and BIRC5. |

  • Proteasome Inhibition: Disrupts PTTG1 degradation, though this may paradoxically stabilize separase activity. |

Pan-Cancer Analysis

  • Expression Patterns: PTTG1 is upregulated in nearly all tumor types, with high expression linked to metastasis and poor prognosis. |

  • Immune Infiltration: Correlates with increased CD8+ T-cell infiltration in certain cancers, suggesting potential for immunotherapy. |

Epigenetic and Mutational Data

  • Genetic Alterations: Rare mutations but frequent amplifications in pituitary and endocrine tumors. |

  • Single-Cell RNAseq: PTTG1 expression peaks during G1/S/G2 phases, aligning with cell cycle progression. |

Target Validation

StrategyOutcomeModel
siRNA Knockdown83.2% reduction in MM tumor burden in vivo.5TGM1 MM cells
Small-Molecule InhibitorsUnder investigation; may disrupt PTTG1-separase interactions or transcriptional activity.Preclinical

Diagnostic Potential

PTTG1’s overexpression in plasma cells and its association with hyperproliferative diseases (e.g., MM) position it as a biomarker for early detection and prognosis. |

Product Specs

Introduction
PTTG1, also known as Securin, plays a crucial role in regulating sister chromatid separation during cell division. This regulatory protein contributes to chromosome stability, the p53/TP53 pathway, and DNA repair. PTTG1 facilitates the transport of separase (a cysteine protease) to the nucleus and inhibits separase's catalytic activity. The Anaphase Promoting Complex (APC) ubiquitinates PTTG1, leading to its degradation by the Proteasome and the subsequent release of separase. During mitosis, PTTG1 prevents premature chromosome segregation by inhibiting Separase/ESPL1, thereby blocking the proteolysis of the cohesin complex. Importantly, PTTG1's function extends beyond inhibition; it is also required for ESPL1 activation. Ubiquitination of PTTG1 at anaphase onset triggers its destruction and liberates ESPL1. PTTG1 possesses two PXXP motifs essential for its transforming and tumorigenic activities and stimulating basic fibroblast growth factor expression. A D box (destruction box) within PTTG1 is crucial for its APC-mediated degradation. The acidic C-terminal region of Securin can act as a transactivation domain. While primarily cytosolic, PTTG1 partially localizes to the nucleus. Overexpression of PTTG1 is observed in various tumors. It exhibits transforming activity in vitro and tumorigenic activity in vivo. Moreover, PTTG1 negatively regulates TP53's transcriptional activity and associated apoptosis.
Description
Recombinant human PTTG1, fused with a 20 amino acid His tag at its N-terminus, is produced in E. coli. This protein is a single, non-glycosylated polypeptide chain consisting of 222 amino acids (residues 1-202) and has a molecular weight of 24.1 kDa. PTTG1 is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The provided PTTG1 solution has a concentration of 1 mg/ml and is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.1 M NaCl, 20% glycerol, 1 mM EDTA, and 0.1 mM PMSF.
Stability
For short-term storage (up to 4 weeks), keep the vial refrigerated at 4°C. For extended storage, freeze the solution at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles to prevent protein degradation.
Purity
The purity of PTTG1 is determined to be greater than 90% by SDS-PAGE analysis.
Synonyms
Securin, Pituitary tumor-transforming gene 1 protein, hPTTG, Tumor-transforming protein 1, Esp1-associated protein, PTTG1, EAP1, PTTG, TUTR1, MGC126883, MGC138276.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MATLIYVDKE NGEPGTRVVA KDGLKLGSGP SIKALDGRSQ VSTPRFGKTF DAPPALPKAT RKALGTVNRA TEKSVKTKGP LKQKQPSFSA KKMTEKTVKA KSSVPASDDA YPEIEKFFPF NPLDFESFDL PEEHQIAHLP LSGVPLMILD EERELEKLFQ LGPPSPVKMP SPPWESNLLQ SPSSILSTLD VELPPVCCDI DI.

Q&A

What is PTTG1 and what is its primary function in normal human cells?

PTTG1 (Pituitary Tumor Transforming Gene 1) encodes mammalian securin, which functions as an inhibitor of separase, a protease essential for the separation of sister chromatids during mitosis and meiosis . In normal tissues, PTTG1 is expressed at low levels, with the only notable exception being testis, where expression is relatively higher than other tissues but still lower than in pituitary adenomas . The primary function of PTTG1 in normal cells is to regulate sister chromatid separation during cell division, which is crucial for maintaining chromosomal stability and preventing aneuploidy. Additionally, PTTG1 is involved in DNA damage repair mechanisms .

How does PTTG1 expression differ between normal and cancerous tissues?

PTTG1 demonstrates significant expression differences between normal and cancerous tissues across multiple cancer types:

Cancer TypePTTG1 Expression PatternAssociated Clinical FeaturesReference
Multiple MyelomaOverexpressed in 36-70% of patientsPoor patient outcomes (HR 2.49)
Esophageal Squamous Cell CarcinomaSignificantly higher than non-cancerous tissuesNegative correlation with age
SeminomaHigh nuclear expressionIncreased invasion capability
Pituitary TumorsOriginally identified as overexpressedAssociated with hyperplasia
Breast CancerMay function as tumor suppressorMice lacking PTTG1 developed mammary tumors

Interestingly, PTTG1 protein levels were found to be down-regulated in human breast tumors, with reduction significantly correlated with tumor grade, suggesting a potential tumor suppressor role in breast tissue .

What experimental models are most effective for studying PTTG1 function?

Various experimental models have proven effective for investigating PTTG1 function:

  • Genetic knockout models: PTTG1-mutant female mice have been used to study mammary epithelial cell development and tumorigenesis, revealing increased proliferation and precocious branching morphogenesis .

  • RNA interference approaches: siRNA knockdown of PTTG1 in cancer cell lines (such as JKT-1 and SEM-1) has been used to evaluate effects on invasion capability and MMP-2 activity .

  • Overexpression systems: Adenovirus expression systems have been employed to deliver PTTG1 into normal human fibroblasts to evaluate its role in early tumorigenesis .

  • In vivo metastasis models: Popliteal lymph node metastasis models in nude mice have been utilized to examine the effect of PTTG1 knockdown on metastatic potential .

  • Cell line panels: Multiple cell lines with different baseline PTTG1 expression (e.g., JKT-1, SEM-1, and TCAM2) allow comparative studies of PTTG1's role in different cellular contexts .

How does PTTG1 function as both an oncogene and tumor suppressor in different contexts?

The dual role of PTTG1 as both oncogene and tumor suppressor represents a significant paradox in cancer biology:

Oncogenic Function:

  • PTTG1 was originally identified as a gene overexpressed in rat pituitary tumors

  • Overexpression is documented in multiple human malignancies including pituitary, colorectal, thyroid, and lung cancers

  • High PTTG1 expression associates with enhanced proliferative capacity, increased tumor grade, and high invasive potential in many cancer types

  • PTTG1 promotes invasiveness through transcriptional activation of matrix metalloproteinases, particularly MMP-2

  • When overexpressed, PTTG1 can cause aneuploidy and genetic instability

Tumor Suppressor Function:

  • In PTTG1-mutant females, mammary epithelial cells showed increased proliferation and precocious branching morphogenesis

  • Mice lacking PTTG1 developed spontaneous mammary tumors

  • In human breast tumors, PTTG1 protein levels were down-regulated, with reduction significantly correlated with tumor grade

  • Molecular changes observed in PTTG1-deficient tissues include up-regulation of progesterone receptor, cyclin D1, and Mmp2, with down-regulation of p21 (Cdkn1a)

This context-dependent function may be explained by tissue-specific interactions, differential subcellular localization, or varying expression levels that determine whether PTTG1 primarily affects chromosome stability, transcriptional regulation, or other cellular processes.

What is the significance of nuclear versus cytoplasmic localization of PTTG1 in cancer progression?

The subcellular localization of PTTG1 appears to be a critical determinant of its function in cancer progression:

Nuclear PTTG1:

  • Associated with an aggressive phenotype in various tumors, including pituitary tumors

  • Correlates with higher migration and invasion capabilities in cancer cell lines

  • Linked to increased MMP-2 levels and activity

  • Functions as a transcription factor regulating genes involved in tumorigenesis

Cytoplasmic PTTG1:

  • Less associated with aggressive phenotypes

  • In TCAM2 cells with predominantly cytoplasmic PTTG1, lower invasive capabilities were observed

  • Overexpression of cytoplasmic PTTG1 may increase MMP-2 protein levels but without significantly increasing MMP-2 activity

The translocation of PTTG1 from cytoplasm to nucleus appears to be regulated by:

  • Interaction with binding partners such as PBF (PTTG1-binding factor)

  • Post-translational modifications, particularly phosphorylation

  • Cell-cycle dependent mechanisms involving CDK1-mediated phosphorylation

Research shows that in JKT-1 cells, PBF overexpression mediated PTTG1 nuclear relocalization, while in TCAM2 cells, PTTG1 remained sequestered in the cytoplasm despite PBF overexpression, suggesting cell-specific mechanisms for regulating PTTG1 localization .

How does PTTG1 overexpression impact chromosomal stability and cellular senescence?

PTTG1 overexpression has profound effects on chromosomal stability and cellular senescence pathways:

Chromosomal Stability:

  • As securin, PTTG1 inhibits separase activation, which is required for sister chromatid separation

  • Overexpression inhibits proper separase function, leading to abnormal nuclei morphologies and chromosome separation defects

  • These abnormalities result in genomic instability and chromosomal aberrations

Cellular Senescence:

  • PTTG1 overexpression in normal human fibroblasts paradoxically inhibits cell proliferation rather than promoting it

  • Several senescence-associated phenotypes emerge, including:

    • Increased SA-β-galactosidase activities

    • Decreased bromodeoxyuridine incorporation

    • Increased SA-heterochromatin foci formation

Mechanistic Pathway:

  • PTTG1 overexpression → Inhibition of separase → Chromosome separation defects

  • Chromosome separation defects → DNA damage response activation

  • DNA damage response → p53 activation → Cellular senescence

This PTTG1-induced senescence is:

  • p53-dependent

  • Telomerase-independent

  • Distinctly different from replicative senescence

This suggests that high PTTG1 levels may initially trigger tumor-suppressive senescence barriers that must be overcome for malignant progression, potentially explaining why additional genetic alterations are required for PTTG1-overexpressing cells to become fully transformed.

What molecular mechanisms underlie PTTG1's role in tumor metastasis and invasion?

PTTG1 promotes metastatic processes through several interconnected molecular mechanisms:

Transcriptional Regulation:

  • PTTG1 functions as a transcriptional activator of matrix metalloproteinases, particularly MMP-2

  • MMP-2 degrades extracellular matrix components, facilitating cancer cell invasion through tissue barriers

  • The transcriptional activity requires nuclear localization of PTTG1

In Vivo Evidence:

  • In a popliteal lymph node metastasis model using highly metastatic HSA/c cells, PTTG1 knockdown significantly reduced metastasis

  • Ratios of metastatic to total dissected popliteal lymph nodes were dramatically lower in PTTG1 siRNA groups compared to controls:

    • 50.0% (5/10) in vehicle control

    • 50.0% (5/10) in non-targeting control

    • 20.0% (2/10) in siRNA P1 group

    • 9.1% (1/11) in siRNA P2 group

Gene Expression Signature:

  • High PTTG1 expression in multiple myeloma correlates with increased expression of cell proliferation-associated genes including:

    • CCNB1, CCNB2 (cyclins)

    • CDK1 (cyclin-dependent kinase)

    • AURKA (Aurora kinase A)

    • BIRC5 (survivin)

    • DEPDC1

  • Knockdown of PTTG1 in 5TGM1 cells decreased expression of Ccnb1, Birc5, and Depdc1 in vitro

Functional Validation:

  • PTTG1 knockdown in 5TGM1 cells significantly reduced MM tumor development in vivo, with an 83.2% reduction in tumor burden at 4 weeks (p<0.0001)

  • PTTG1 expression is part of a 17-gene molecular signature capable of predicting tumor metastasis

These findings collectively demonstrate that PTTG1 facilitates metastasis through transcriptional activation of invasion-promoting genes, cell cycle regulators, and anti-apoptotic factors.

What are the most effective techniques for studying PTTG1 localization and its functional consequences?

Effective techniques for studying PTTG1 localization include:

Subcellular Fractionation:

  • Separation of nuclear and cytoplasmic fractions followed by western blotting to quantify PTTG1 distribution

  • Allows biochemical assessment of PTTG1 compartmentalization

Immunofluorescence Microscopy:

  • Direct visualization of PTTG1 subcellular localization using specific antibodies

  • Can be combined with co-localization studies using markers for specific organelles

Reporter Constructs:

  • Tagged PTTG1 constructs (FLAG-PTTG1, GFP-PTTG1) to track localization in live cells

  • Useful for dynamic studies of translocation

Functional Correlation Studies:

  • Correlating localization with functional readouts:

    • Invasion assays (Transwell/Matrigel)

    • MMP-2 zymography to assess enzymatic activity

    • Cell proliferation assays

    • Chromosomal stability assessments

Phosphorylation State Analysis:

  • Phospho-specific antibodies or mass spectrometry to detect post-translational modifications that influence localization

  • Site-directed mutagenesis of phosphorylation sites to study their impact on localization and function

Protein-Protein Interaction Studies:

  • Co-immunoprecipitation to identify interacting partners that influence localization (e.g., PBF)

  • Proximity ligation assays to verify interactions in situ

For maximum insight, combining multiple approaches is recommended to establish clear correlations between PTTG1 localization patterns and functional outcomes in different cellular contexts.

How can researchers reconcile contradictory findings about PTTG1's role in different cancer types?

Reconciling contradictory findings about PTTG1 requires systematic approaches:

1. Context-Specific Analysis:

  • Conduct parallel studies across multiple tissue types using identical methodologies

  • Establish tissue-specific baseline expression levels and functional networks

  • Consider the broader molecular context, including p53 status, which affects PTTG1-induced senescence

2. Dose-Response Relationships:

  • Investigate whether PTTG1 exhibits biphasic effects depending on expression level

  • Use inducible expression systems with titratable control to test functional outcomes at different expression levels

3. Temporal Dynamics:

  • Study PTTG1's role at different stages of cancer progression

  • Differentiate between early effects (potentially tumor-suppressive through senescence induction) and late effects (potentially oncogenic through aneuploidy and MMP activation)

4. Interaction Network Mapping:

  • Identify tissue-specific binding partners that modify PTTG1 function

  • Perform comparative interactomics across different cell types

5. Integrated Multi-Omics Approach:

  • Combine genomics, transcriptomics, proteomics, and functional studies

  • Example methodology table:

ApproachTechnologyOutcome MeasureAdvantage
TranscriptomicsRNA-seq after PTTG1 modulationDifferentially expressed genesIdentifies context-specific targets
ProteomicsIP-MSPTTG1 interacting proteinsReveals tissue-specific complexes
ChIP-seqPTTG1 chromatin bindingDirect transcriptional targetsDistinguishes direct vs. indirect effects
CRISPR screeningSynthetic lethalityGenetic dependenciesIdentifies context-specific vulnerabilities

6. Standardized Reporting:

  • Clearly document experimental conditions, cell passage numbers, and expression levels

  • Report subcellular localization data alongside functional outcomes

By implementing these approaches, researchers can develop a unified model that explains PTTG1's seemingly contradictory roles across different cancer contexts.

What animal models are most suitable for investigating PTTG1's role in tumorigenesis?

Several animal models have proven valuable for investigating PTTG1's role in tumorigenesis:

Genetic Knockout Models:

  • PTTG1-mutant mice exhibit tissue-specific phenotypes, including increased mammary epithelial cell proliferation and precocious branching morphogenesis

  • These mice develop spontaneous mammary tumors, supporting PTTG1's tumor-suppressive role in breast tissue

  • PTTG1 deletion provides protection against Rb haploinsufficiency-induced pituitary tumorigenesis, suggesting context-dependent functions

Transgenic Overexpression Models:

  • Transgenic overexpression of human PTTG1 in mouse pituitary causes hyperplasia and adenoma

  • Tissue-specific promoters can target PTTG1 overexpression to particular organs of interest

Metastasis Models:

  • Popliteal lymph node metastasis model in nude mice provides a quantifiable system for assessing PTTG1's impact on metastatic potential

  • This model allows for direct injection of siRNA to maintain knockdown effects throughout the experiment

Xenograft Models:

  • 5TGM1 myeloma cells with PTTG1 knockdown showed 83.2% reduction in tumor burden when injected into mice

  • Cell line-derived xenografts allow assessment of both primary tumor growth and metastatic potential

Experimental Considerations:

  • For studying dual oncogenic/tumor-suppressive roles, conditional knockout/knockin models are preferable

  • Tissue-specific and inducible systems allow temporal control of PTTG1 expression

  • Combined genetic models (e.g., PTTG1 modulation in p53-null background) help decipher pathway interactions

The choice of model should be guided by the specific aspect of PTTG1 biology under investigation, with consideration of tissue context and baseline expression levels.

How reliably does PTTG1 expression correlate with patient outcomes in different cancer types?

PTTG1 expression shows variable but significant correlations with patient outcomes across cancer types:

Multiple Myeloma:

  • High PTTG1 expression significantly associated with poor patient outcomes

  • Hazard ratio of 2.49 (95% CI 1.28 to 4.86; p = 0.0075) in patients from the Total Therapy 2 trial

  • The quartile with highest PTTG1 expression had significantly poorer survival compared to remaining patients

Esophageal Squamous Cell Carcinoma (ESCC):

Breast Cancer:

  • Contrary to many other cancers, PTTG1 protein levels were down-regulated in human breast tumors

  • This reduction significantly correlated with increasing tumor grade, suggesting a potential tumor suppressor role

General Cancer Metastasis:

  • PTTG1 expression represents one of 17 genes that form a molecular signature capable of predicting tumor metastasis

The reliability of PTTG1 as a prognostic marker appears to be:

  • Most robust in multiple myeloma

  • Suggestive but not definitive in ESCC

  • Complex and potentially inverse in breast cancer

This suggests that PTTG1 expression should be interpreted in a cancer type-specific context and potentially in combination with other markers for optimal prognostic value.

What is the role of PTTG1 in therapy resistance and how might it be targeted?

Though the provided search results don't directly address therapy resistance mechanisms, we can extract insights about potential therapeutic approaches based on PTTG1 biology:

Potential PTTG1-Related Resistance Mechanisms:

  • Chromosomal instability induced by PTTG1 overexpression may accelerate the acquisition of therapy-resistant mutations

  • PTTG1-mediated upregulation of survival factors like BIRC5 (survivin) could confer resistance to apoptosis-inducing therapies

  • Nuclear localization of PTTG1 promotes invasion and metastasis, potentially contributing to treatment failure

Therapeutic Targeting Approaches:

  • RNA Interference:

    • siRNA targeting of PTTG1 has shown efficacy in reducing invasion capability and MMP-2 activity in cancer cell lines

    • In vivo delivery of PTTG1 siRNA significantly reduced lymph node metastasis in animal models

    • PTTG1 knockdown in 5TGM1 cells decreased tumor burden by 83.2% in vivo

  • Nuclear Localization Inhibition:

    • Targeting the nuclear translocation of PTTG1 could be effective in cancers where nuclear PTTG1 drives aggressive behavior

    • Inhibiting interactions with nuclear transport facilitators like PBF represents a potential approach

    • Modulating the post-translational modifications (particularly phosphorylation) that regulate PTTG1 localization

  • Synthetic Lethality:

    • Exploiting the chromosomal instability induced by PTTG1 overexpression by combining with agents that target cells with DNA damage

    • Cells with PTTG1-induced chromosomal abnormalities might be more sensitive to PARP inhibitors or checkpoint inhibitors

  • Context-Specific Approaches:

    • In breast cancer, where PTTG1 may function as a tumor suppressor, strategies to restore or enhance PTTG1 function might be beneficial

    • In multiple myeloma and other cancers where PTTG1 is clearly oncogenic, direct inhibition approaches are warranted

These therapeutic considerations must account for the context-dependent roles of PTTG1 across different cancer types and stages of progression.

How do post-translational modifications regulate PTTG1 function and localization?

Post-translational modifications (PTMs) play crucial roles in regulating PTTG1 function and subcellular localization:

Phosphorylation:

  • Specific phosphorylation of PTTG1 has been demonstrated to be responsible for its nuclear localization

  • Cyclin-dependent Kinase 1 (CDK1) mediates PTTG1 phosphorylation, which affects its Golgi membrane localization

  • These phosphorylation events likely create binding sites for nuclear transport proteins or mask cytoplasmic retention signals

Other Potential PTMs:

  • While not explicitly detailed in the provided search results, other PTMs that commonly regulate protein localization and function may affect PTTG1:

    • Ubiquitination (affecting protein stability)

    • SUMOylation (often affecting nuclear-cytoplasmic transport)

    • Acetylation (potentially affecting DNA binding capabilities)

Experimental Approaches to Study PTTG1 PTMs:

  • Mass spectrometry to identify specific modification sites

  • Phospho-specific antibodies to track modified PTTG1 in different cellular compartments

  • Site-directed mutagenesis of potential modification sites to create phospho-mimetic or phospho-dead variants

  • Real-time imaging of fluorescently tagged PTTG1 to track localization dynamics in response to stimuli

Regulatory Interactions:

  • PTTG1-binding factor (PBF) mediates PTTG1 nuclear relocalization in some cell types (e.g., JKT-1) but not in others (e.g., TCAM2)

  • This suggests that PTMs may regulate protein-protein interactions critical for PTTG1 trafficking

Understanding these regulatory mechanisms could provide new opportunities for therapeutic intervention by targeting specific enzymes responsible for PTTG1 modifications or by developing compounds that mimic or block critical PTM sites.

What is the relationship between PTTG1 and the DNA damage response pathway?

PTTG1 has a complex relationship with the DNA damage response (DDR) pathway:

PTTG1 Overexpression Activates DDR:

  • When overexpressed in normal human fibroblasts, PTTG1 inhibits proper sister chromatid separation

  • This leads to chromosomal instability and abnormal nuclei morphologies

  • These genomic abnormalities trigger activation of the DNA damage response pathway

  • The activated DDR subsequently induces p53-dependent cellular senescence

PTTG1 in DNA Repair:

  • Beyond its role in causing DNA damage through chromosomal instability, PTTG1 is also directly involved in DNA damage repair mechanisms

  • This suggests a potential feedback loop where PTTG1 may both cause and respond to DNA damage

P53 Dependency:

  • The PTTG1-induced senescence observed in normal human fibroblasts is p53-dependent

  • This indicates that functional p53 is required for cells to activate the senescence program in response to PTTG1-induced chromosomal instability

  • In cancer cells with p53 mutations, this protective senescence response may be compromised, potentially explaining how elevated PTTG1 can drive cancer progression in these contexts

Telomerase Independence:

  • Unlike replicative senescence, PTTG1-induced senescence is telomerase-independent

  • This distinguishes the DDR activation by PTTG1 from telomere erosion-associated DNA damage

The relationship between PTTG1 and DDR represents a potential vulnerability that could be exploited therapeutically, particularly in cancers with high PTTG1 expression but intact DDR pathways.

How does PTTG1 interact with other cell cycle regulators to control proliferation?

PTTG1 interacts with multiple cell cycle regulators to influence proliferation:

Direct Cell Cycle Interactions:

  • As securin, PTTG1 inhibits separase, preventing premature sister chromatid separation during mitosis

  • This function is critical for maintaining chromosomal stability during cell division

  • Overexpression inhibits proper separase activation, leading to abnormal chromosome segregation

Transcriptional Regulation:

  • PTTG1 directly or indirectly regulates transcription of several cell cycle genes:

    • Cdkn1a (p21) - downregulated in PTTG1-mutant mammary epithelial cells

    • Cyclin D1 - upregulated in PTTG1-mutant mammary epithelial cells

    • CCNB1, CCNB2 (cyclins) - expression correlated with PTTG1 in multiple myeloma

    • CDK1 - expression correlated with PTTG1 in multiple myeloma

    • AURKA (Aurora kinase A) - expression correlated with PTTG1 in multiple myeloma

Functional Cell Cycle Effects:

  • PTTG1 knockdown in 5TGM1 cells decreased cellular proliferation without affecting cell cycle distribution or viability

  • In normal human fibroblasts, PTTG1 overexpression paradoxically inhibited cell proliferation and induced senescence

  • In mammary epithelial cells of PTTG1-mutant females, increased proliferation and precocious branching morphogenesis were observed

Regulatory Network:

  • CDK1 can phosphorylate PTTG1, affecting its localization and potentially creating a feedback loop in cell cycle regulation

  • The p53-dependency of PTTG1-induced senescence suggests interplay between PTTG1 and the p53 tumor suppressor pathway

These interactions create a complex regulatory network where PTTG1's effects on proliferation depend on:

  • Cell type and tissue context

  • Expression level

  • Subcellular localization

  • Status of other cell cycle regulators (particularly p53)

  • Post-translational modification state

Product Science Overview

Introduction

Pituitary Tumor-Transforming Protein 1 (PTTG1), also known as securin, is a protein encoded by the PTTG1 gene in humans. It plays a crucial role in cell cycle regulation, particularly in the separation of sister chromatids during mitosis. PTTG1 has garnered significant attention due to its involvement in various cancers and its potential as a therapeutic target.

Discovery and Structure

PTTG1 was first identified in the context of pituitary tumors, where it was found to be overexpressed. The protein consists of 202 amino acids and contains a PXXP motif, which is crucial for its interaction with other proteins. The structure of PTTG1 allows it to bind to and inhibit separase, an enzyme responsible for cleaving cohesin complexes that hold sister chromatids together.

Biological Functions

PTTG1 is a multifunctional protein with several roles in cellular processes:

  • Cell Cycle Regulation: PTTG1 inhibits separase, preventing premature separation of sister chromatids. This ensures proper chromosome segregation during cell division.
  • Transcriptional Regulation: PTTG1 can act as a transcriptional activator, influencing the expression of various genes involved in cell proliferation and survival.
  • DNA Repair: PTTG1 is implicated in DNA repair mechanisms, contributing to genomic stability.
  • Tumorigenesis: Overexpression of PTTG1 is associated with tumorigenesis. It promotes cell proliferation, inhibits apoptosis, and enhances angiogenesis, making it a key player in cancer progression.
Clinical Significance

PTTG1 is overexpressed in various cancers, including pituitary adenomas, breast cancer, gastrointestinal cancers, leukemia, lymphoma, and lung cancer . Its overexpression is often correlated with poor prognosis and increased tumor aggressiveness. Due to its role in tumorigenesis, PTTG1 is considered a potential biomarker for cancer diagnosis and prognosis.

Therapeutic Potential

Given its involvement in cancer, PTTG1 is a promising target for therapeutic interventions. Strategies to inhibit PTTG1 expression or function could potentially suppress tumor growth and improve patient outcomes. Research is ongoing to develop specific inhibitors and immunotherapies targeting PTTG1 .

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.