PA2G4 Human

Proliferation-associated protein 2G4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

The PA2G4 gene spans 8.4 kb and comprises ten exons, producing two splice variants:

  • p48 EBP1 (394 amino acids): Transcription initiates at the first ATG codon.

  • p42 EBP1 (340 amino acids): Generated via alternative splicing, omitting exon 2b and initiating translation at the third ATG codon .

Featurep48 EBP1p42 EBP1
Molecular Weight48 kDa42 kDa
N-terminal DomainContains 1.5 α-helicesLacks N-terminal helices
StabilityStableUbiquitin-proteasome degradation
Cellular LocalizationNucleus and cytoplasmPrimarily cytoplasm

Structurally, PA2G4 adopts a methionine aminopeptidase (MetAP) fold but lacks catalytic activity. The lysine-rich C-terminal region (residues 364–373) mediates RNA/DNA binding, critical for interactions with viral internal ribosome entry sites (IRES) and transcriptional regulation .

Isoform-Specific Functions in Cancer

PA2G4 isoforms exhibit opposing roles in tumorigenesis, acting as oncogenes or tumor suppressors depending on context:

p48 EBP1: Oncogenic Roles

  • Mechanisms: Binds HDM2 to promote p53 degradation , stabilizes MYCN in neuroblastoma , and activates PI3K/AKT signaling .

  • Clinical Relevance: Overexpressed in glioblastoma (GBM), acute myeloid leukemia (AML), and oral squamous cell carcinoma (OSCC), correlating with poor prognosis .

p42 EBP1: Tumor-Suppressive Roles

  • Mechanisms: Degrades PI3K via HSP70/CHIP-mediated ubiquitination , inhibits AKT, and suppresses E2F-1 via RB interaction .

  • Clinical Relevance: Downregulated in prostate cancer, GBM, and non-small cell lung cancer (NSCLC) .

Cancer Typep48 EBP1 Rolep42 EBP1 Role
GlioblastomaPromotes growth via p53 inhibition Suppressed; loss correlates with progression
Prostate CancerLow expressionBinds androgen receptor (AR) to repress PSA
NeuroblastomaStabilizes MYCN oncoprotein Not detected

Developmental and Regulatory Roles

PA2G4 is essential in embryogenesis and neural development:

  • Neural Crest Development: Interacts with Six1 to regulate neural border zone genes. Knockdown in Xenopus reduces otocyst size and disrupts cranial placode formation .

  • Stem Cell Differentiation: p48 EBP1 maintains pluripotency by binding DPPA4 in embryonic stem cells .

Therapeutic Targeting

PA2G4-p48 is a promising target in MYCN-driven cancers:

  • WS6 Analogues: Small molecules (e.g., SVI-3567, SVI-5333) inhibit PA2G4-MYCN interaction, reducing tumor growth in neuroblastoma (IC50: 23–30 µM) .

  • HDM2 Inhibitors: Block p48-mediated p53 degradation, restoring tumor suppressor activity .

CompoundTargetMechanismTherapeutic Window
WS6PA2G4-MYCN interfaceDisrupts oncogenic feedback loop2.1
SVI-5333PA2G4 lysine-rich domainReduces MYCN stability3.6

Key Interactions and Pathways

PA2G4 integrates multiple signaling networks:

  • ErbB3 Receptor: Modulates growth signals via cytoplasmic interaction .

  • Androgen Receptor (AR): p42 EBP1 represses AR-dependent transcription in prostate cancer .

  • PI3K/AKT: p42 inhibits, while p48 activates this pathway .

Future Directions

Research gaps include isoform-specific structural studies and clinical validation of PA2G4 inhibitors. Targeting the lysine-rich RNA-binding domain or isoform-specific post-translational modifications (e.g., p42 sumoylation ) could yield precision therapies.

Product Specs

Introduction
PA2G4, a member of the peptidase M24C family, is an RNA-binding protein with roles in cellular proliferation and differentiation. It participates in ribosome assembly and rRNA processing as a component of pre-ribosomal ribonucleoprotein complexes. PA2G4 interacts with ErbB-3, potentially modulating Neuregulin-1 signaling in the ErbB-3 pathway. Furthermore, it acts as a transcriptional co-repressor of androgen receptor-regulated and cell cycle regulatory genes by interacting with histone deacetylases. PA2G4 is implicated in growth inhibition and differentiation induction in human cancer cells. Additionally, it mediates cap-independent translation of specific viral internal ribosomal entry sites (IRESs). PA2G4 associates with mature 28S, 18S, and 5.8S rRNAs, various rRNA precursors, and possibly U3 small nucleolar RNA.
Description
PA2G4, produced in E. coli, is a non-glycosylated polypeptide chain containing 402 amino acids (with amino acids 1-394) and a molecular mass of 44.8 kDa. It is expressed with an 8-amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The PA2G4 protein is supplied in a solution containing 20mM Tris-HCl buffer at pH 8.0 and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), store the protein at 4°C. For long-term storage, freeze the protein at -20°C. It is recommended to add a carrier protein such as HSA or BSA (0.1%) for extended storage. Avoid repeated freezing and thawing.
Purity
The purity of the protein is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Proliferation-associated protein 2G4, Cell cycle protein p38-2G4 homolog, hG4-1, ErbB3-binding protein 1, PA2G4, EBP1, p38-2G4.
Source
Escherichia Coli.
Amino Acid Sequence
MSGEDEQQEQ TIAEDLVVTK YKMGGDIANR VLRSLVEASS SGVSVLSLCE KGDAMIMEET GKIFKKEKEM KKGIAFPTSI SVNNCVCHFS PLKSDQDYIL KEGDLVKIDL GVHVDGFIAN VAHTFVVDVA QGTQVTGRKA DVIKAAHLCA EAALRLVKPG NQNTQVTEAW NKVAHSFNCT PIEGMLSHQL KQHVIDGEKT IIQNPTDQQK KDHEKAEFEV HEVYAVDVLV SSGEGKAKDA GQRTTIYKRD PSKQYGLKMK TSRAFFSEVE RRFDAMPFTL RAFEDEKKAR MGVVECAKHE LLQPFNVLYE KEGEFVAQFK FTVLLMPNGP MRITSGPFEP DLYKSEMEVQ DAELKALLQS SASRKTQKKK KKKASKTAEN ATSGETLEEN EAGDLEHHHH HH.

Q&A

What is the basic structure and organization of the human PA2G4 gene?

The human PA2G4 gene encodes an RNA-binding protein involved in growth regulation. It contains three in-frame ATG initiation codons, which through differential splicing produce two major isoforms: PA2G4-p42 (340 amino acids) and PA2G4-p48 (394 amino acids). The gene is classified as protein-coding with the Entrez Gene ID of 5036. Six pseudogenes related to PA2G4 have been identified on chromosomes 3, 6, 9, 18, 20, and X .

Structurally, the PA2G4 protein possesses several functional domains, including an amphipathic helical domain (amino acids 204-246) that facilitates DNA and protein interactions. The protein also contains lysine-rich regions and multiple protein-protein interaction sites that are crucial for its diverse cellular functions .

What are the key differences between the two major PA2G4 isoforms?

The two PA2G4 isoforms differ significantly in their cellular functions and localization:

FeaturePA2G4-p48PA2G4-p42
Length394 amino acids340 amino acids
Expression levelPredominant form in mammalian cellsExpressed at relatively low levels
Cellular localizationBoth nucleus and cytoplasmPrimarily cytoplasmic
Role in cancerPro-tumorigenic (oncogenic)Tumor suppressive
Transcription startFirst ATG codonThird ATG codon

These isoforms often have opposing roles depending on cellular context. The structural differences between these isoforms translate into distinct functional outcomes, with PA2G4-p48 generally promoting tumorigenesis while PA2G4-p42 acts as a tumor suppressor .

How conserved is PA2G4 across species?

PA2G4 shows strong evolutionary conservation. The human PA2G4 protein shares significant similarity with the murine p38-2G4 protein. Structural comparisons between human and murine PA2G4-p48 reveal a highly conserved structural fold with a root mean square deviation (RMSD) across the α-carbons of only 0.44 Å, indicating nearly identical three-dimensional structures .

This high degree of conservation suggests fundamental biological importance across mammalian species, particularly for functions related to cellular growth regulation and ribosomal processing.

What structural data is currently available for PA2G4, and what do these structures reveal about its function?

Three major protein structures of PA2G4 have been published to date:

  • A 2.5 Å resolution X-ray crystal structure of murine PA2G4-p48 (PDB entry 2V6C) derived from an N- and C-terminally truncated construct (amino acids 8-360)

  • A higher-resolution (1.6 Å) X-ray crystal structure of human PA2G4-p48 (PDB entry 2Q8K) with electron density observed between residues 7 and 362

  • A cryo-EM structure of human PA2G4-p48 in complex with the human 80S ribosome (PDB entry 6SXO) at 3.3-8 Å resolution, notable for resolving the most C-terminal 33 amino acids

Additionally, a small 20-amino acid fragment of PA2G4 in complex with HNF4α was solved (PDB entry 6CHT), though only 7 amino acids (AELKALL) were resolved .

These structures reveal important functional domains including nucleic acid binding regions, protein-protein interaction surfaces, and structural features that explain PA2G4's roles in ribosome assembly and transcriptional regulation.

How do the structural features of PA2G4 isoforms explain their opposing functions in cancer?

The structural differences between PA2G4 isoforms contribute to their contrasting roles in cancer:

The PA2G4-p48 isoform contains an extended N-terminal region (54 amino acids) absent in PA2G4-p42. This region may contain binding sites for oncogenic partners or nuclear localization signals that enable distinct protein interactions.

The C-terminal domain (residues 300-372) is critical for DNA binding and interaction with tumor suppressor proteins like Rb. This domain is present in both isoforms but may function differently based on the presence or absence of the N-terminal region and associated post-translational modifications .

Lysine-rich regions in PA2G4 serve as sites for post-translational modifications that can dramatically alter protein function. These modifications likely occur differently between the isoforms, directing them toward either oncogenic or tumor-suppressive pathways .

What post-translational modifications regulate PA2G4 function?

PA2G4 undergoes various post-translational modifications that regulate its activity and cellular localization. Key modifications include:

  • Phosphorylation events that can alter protein-protein interactions, particularly with partners like Akt and HDM2

  • Sumoylation which may affect nuclear-cytoplasmic shuttling

  • Ubiquitination that regulates protein stability and degradation

These modifications differ between the isoforms and contribute to their distinct functional outcomes. For instance, differential phosphorylation could determine whether PA2G4 activates oncogenic pathways or tumor-suppressive mechanisms .

What are the key protein-protein interactions of PA2G4 that influence cancer progression?

PA2G4 interacts with numerous proteins that influence cancer progression:

  • ErbB3 receptor: PA2G4 binds to the cytoplasmic domain of ErbB3, potentially modulating growth regulatory signals .

  • Retinoblastoma (Rb) protein and Sin3A: PA2G4 interacts indirectly with E2F1 at promoter elements by binding to a complex including Rb and Sin3A, affecting cell cycle regulation .

  • Histone deacetylases (HDACs): PA2G4 binds to HDAC2 both in vitro and in vivo, with this interaction being critical for its transcriptional repression activity .

  • Akt: PA2G4 strongly binds protein kinase B (Akt) and suppresses apoptosis. Phosphorylated nuclear Akt interacts with PA2G4 and enhances its antiapoptotic action .

  • HDM2 and p53: PA2G4 binds to HDM2, enhancing HDM2-p53 association and promoting p53 polyubiquitination and degradation, thereby decreasing p53 activity .

  • MYCN: Interactions with MYCN appear to be significant for PA2G4's oncogenic functions and represent potential therapeutic targets .

How does PA2G4 participate in RNA processing and translational regulation?

PA2G4 is involved in multiple aspects of RNA metabolism:

The protein is present in pre-ribosomal ribonucleoprotein complexes and participates in ribosome assembly and the regulation of intermediate and late steps of rRNA processing . This function is supported by the cryo-EM structure showing PA2G4 in complex with the 80S ribosome .

PA2G4 contains an amphipathic helical domain (amino acids 204-246) that facilitates direct interaction with nucleic acids. The C-terminal domain functions as an important DNA-binding domain .

Through its involvement in ribosome maturation and potential direct interactions with mRNAs, PA2G4 can influence translational output and thus affect cellular growth and differentiation programs. These functions may be differentially regulated between the two major isoforms .

What is known about the transcriptional regulatory functions of PA2G4?

PA2G4 serves as a transcriptional co-repressor for multiple genes:

It functions as a co-repressor of androgen receptor-regulated genes through interactions with histone deacetylases (HDACs). HDAC inhibitors like trichostatin A can significantly reduce PA2G4-mediated repression .

The protein interacts with E2F1 promoter elements by binding to a complex of nuclear proteins, including retinoblastoma (Rb) protein and Sin3A, thus participating in cell cycle regulation .

PA2G4's C-terminal domain (residues 300-372) is necessary for Rb binding, which is important for transcriptional repression in Rb-positive cells. PA2G4 mutants lacking this Rb-binding domain lose their ability to repress transcription of cell cycle genes like cyclin E .

How does PA2G4 expression correlate with prognosis in different cancer types?

PA2G4 expression levels correlate with prognosis in various human cancers:

Multivariate analysis identified PA2G4 expression as an independent prognostic indicator for survival in NPC patients, with elevated expression associated with unfavorable outcomes .

Beyond NPC, PA2G4 expression has prognostic value in neuroblastoma, cervical, brain, breast, prostate, pancreatic, and hepatocellular cancers, though the prognostic significance may depend on which isoform is predominantly expressed in the specific cancer type .

What is the evidence supporting PA2G4 as a potential therapeutic target in cancer?

Several lines of evidence support PA2G4 as a potential therapeutic target:

The oncogenic PA2G4-p48 isoform promotes tumorigenesis across multiple cancer types. Studies suggest targeting protein-protein interaction sites essential for the oncogenic role of PA2G4, such as its interaction with MYCN, could be therapeutically beneficial .

A small molecule inhibitor called WS6 has been identified that interacts with PA2G4, suggesting the feasibility of pharmacological targeting .

Potential approaches include targeting protein partners like FBXW1, which interacts with both isoforms but elicits alternate functional effects in cells .

The challenge in therapeutic development lies in selectively targeting the oncogenic PA2G4-p48 isoform while sparing the tumor-suppressive PA2G4-p42 isoform. Structure-based drug design campaigns must take into account isoform-specific features to achieve this selectivity .

How is PA2G4 expression dysregulated in nasopharyngeal carcinoma?

Research indicates significant dysregulation of PA2G4 in nasopharyngeal carcinoma (NPC):

Immunohistochemical analysis of 201 NPC cases and 45 normal nasopharyngeal tissues revealed that PA2G4 protein expression was significantly higher in NPC tissues compared to non-cancerous tissues (p=0.005) .

Microarray data analysis showed that PA2G4 levels were significantly increased in both NPC cell lines and pooled NPC tissues compared with normal nasopharyngeal tissues .

This overexpression correlates with multiple adverse clinical parameters:

  • Larger tumor size (T classification) (p<0.001)

  • Lymph node metastasis (N classification) (p<0.001)

  • Distant metastasis (p=0.029)

  • Advanced clinical stage (p<0.001)

Stratified analysis indicated that high PA2G4 expression was associated with worse survival specifically in advanced clinical stages (III-IV) .

What are the recommended approaches for studying isoform-specific functions of PA2G4?

When studying isoform-specific functions of PA2G4, researchers should consider:

Isoform-Specific Antibodies: Develop and validate antibodies that specifically recognize either PA2G4-p42 or PA2G4-p48 for proper discrimination in experimental settings. Western blotting with appropriate controls is essential to confirm isoform specificity.

Expression Constructs: Use expression vectors containing the specific coding sequence for either PA2G4-p42 or PA2G4-p48. These should be validated by sequencing and protein expression confirmation.

RNA Interference and CRISPR-Cas9: Design targeting strategies that can selectively knockdown or knockout specific isoforms based on their unique sequences. For PA2G4, targeting the N-terminal region present only in the p48 isoform can provide isoform specificity.

Mass Spectrometry: Employ proteomic approaches to identify isoform-specific post-translational modifications and protein-protein interactions.

Genetically Engineered Mouse Models: Consider developing mouse models that express only one isoform to assess its biological impact in normal and cancer cells. As suggested in the literature, PA2G4-p48 knockout mice could be valuable in determining whether depletion of this isoform delays or prevents tumor growth .

What structural biology techniques have been most informative for PA2G4 characterization?

Multiple structural biology techniques have advanced our understanding of PA2G4:

X-ray Crystallography: Provided high-resolution structures of both murine (2.5 Å) and human (1.6 Å) PA2G4-p48, revealing detailed information about protein fold and potential functional sites .

Cryo-Electron Microscopy (Cryo-EM): Resolved PA2G4-p48 in complex with the human 80S ribosome at 3.3-8 Å resolution, uniquely revealing the most C-terminal 33 amino acids not visible in crystal structures and providing insights into PA2G4's role in ribosome function .

Protein-Protein Complex Crystallography: Structures like the PA2G4-HNF4α complex provide insights into specific interaction interfaces, though sometimes with limited resolution .

For future studies, integrating these techniques with hydrogen-deuterium exchange mass spectrometry (HDX-MS), nuclear magnetic resonance (NMR), and small-angle X-ray scattering (SAXS) could further elucidate dynamic aspects of PA2G4 structure and function, particularly differences between isoforms.

What experimental models are most appropriate for investigating PA2G4's role in cancer progression?

Several experimental models have proven valuable for investigating PA2G4's role in cancer:

Cell Line Models: Cancer cell lines with differential expression of PA2G4 isoforms can be used to study isoform-specific effects on proliferation, apoptosis, and gene expression. Manipulation of PA2G4 levels through overexpression or knockdown provides insights into its functional impact.

Patient-Derived Xenografts (PDX): These models maintain the heterogeneity of human tumors and can better reflect the clinical relevance of PA2G4 expression patterns observed in patients.

Genetically Engineered Mouse Models: As suggested in the literature, PA2G4-p48 knockout mice crossed with murine cancer models could determine whether depletion of this isoform affects tumor development and progression .

Tissue Microarrays: For clinical correlation studies, tissue microarrays with large cohorts of patient samples allow for robust assessment of PA2G4 expression in relation to clinicopathological features and survival outcomes, as demonstrated in nasopharyngeal carcinoma research .

3D Organoid Models: These increasingly popular models provide a more physiologically relevant environment than traditional 2D cell culture and could offer insights into PA2G4's role in tumor microenvironment interactions.

What are the critical unresolved questions about PA2G4 function in cancer biology?

Several important questions about PA2G4 remain unanswered:

  • How do the two PA2G4 isoforms compete for binding partners, and how does this competition affect cellular outcomes? The fact that PA2G4-p42 acts as a tumor suppressor through its C-terminal region (which is conserved in PA2G4-p48) suggests complex competitive binding dynamics that require further investigation .

  • What determines the relative expression of PA2G4 isoforms in different tissues and cancer types? Understanding the splicing regulation mechanisms could provide insights into tissue-specific functions.

  • How do post-translational modifications differ between the isoforms, and how do these modifications direct them toward either oncogenic or tumor-suppressive functions?

  • What is the full complement of proteins that interact with each PA2G4 isoform, and how do these interaction networks differ between normal and cancer cells?

  • What is the mechanism by which PA2G4 contributes to ribosome assembly and rRNA processing, and how does this function relate to its roles in transcriptional regulation?

How might targeting PA2G4 be integrated into combination therapy approaches?

Potential combination therapy approaches involving PA2G4 include:

With HDAC Inhibitors: Since PA2G4 functions partly through recruitment of histone deacetylases, combining PA2G4 inhibitors with established HDAC inhibitors might synergistically enhance anti-tumor effects.

With ErbB Pathway Inhibitors: Given PA2G4's interaction with the ErbB3 receptor, combination with ErbB family tyrosine kinase inhibitors could target multiple nodes in this signaling pathway.

With p53 Pathway Modulators: PA2G4 modulates p53 levels through interaction with HDM2. Combining PA2G4 inhibitors with drugs that target the p53-HDM2 interaction could potentially restore p53 tumor suppressor function.

Isoform-Specific Targeting: Developing therapeutics that specifically inhibit the oncogenic PA2G4-p48 while sparing or even enhancing PA2G4-p42 function could provide a more refined approach to targeting PA2G4 in cancer.

Biomarker-Guided Therapy: Using PA2G4 expression patterns (total and isoform-specific) as biomarkers could help identify patients most likely to benefit from PA2G4-targeted therapies or specific combination approaches.

What emerging technologies could advance our understanding of PA2G4 biology?

Several cutting-edge technologies could significantly advance PA2G4 research:

Single-Cell Analysis: Single-cell RNA sequencing and proteomics could reveal cell-to-cell variability in PA2G4 isoform expression and function within heterogeneous tumor samples.

Spatial Transcriptomics and Proteomics: These techniques could map PA2G4 expression patterns within the complex tumor microenvironment, providing insights into its role in tumor-stroma interactions.

CRISPR Screening: Genome-wide CRISPR screens in the context of PA2G4 manipulation could identify synthetic lethal interactions and novel functional connections.

Ribosome Profiling: This technique could elucidate PA2G4's specific roles in translational regulation by identifying the mRNAs whose translation is affected by PA2G4 alterations.

Cryo-Electron Tomography: This emerging method could provide structural insights into PA2G4's interactions within larger macromolecular complexes in a more native cellular context.

AI-Driven Structure Prediction and Drug Design: Technologies like AlphaFold2 could help predict the full structure of both PA2G4 isoforms, including regions not resolved in current experimental structures, facilitating more effective isoform-specific drug design.

Product Science Overview

Gene and Protein Structure

The PA2G4 gene is located on chromosome 12 and is a protein-coding gene. It encodes an RNA-binding protein that is involved in growth regulation. The protein is present in pre-ribosomal ribonucleoprotein complexes and may be involved in ribosome assembly and the regulation of intermediate and late steps of rRNA processing .

Function and Mechanism

PA2G4 interacts with the cytoplasmic domain of the ErbB3 receptor, contributing to the transduction of growth regulatory signals. It also acts as a transcriptional co-repressor of androgen receptor-regulated genes and other cell cycle regulatory genes through its interactions with histone deacetylases . This protein has been implicated in growth inhibition and the induction of differentiation of human cancer cells .

Expression and Localization

PA2G4 is ubiquitously expressed in various tissues, with cytoplasmic localization. It is involved in several pathways, including the innate immune system and the coregulation of androgen receptor activity . The protein’s expression and localization suggest its involvement in fundamental cellular processes.

Clinical Significance

Mutations or dysregulation of the PA2G4 gene have been associated with several diseases, including trichodentoosseous syndrome and prostate cancer . Its role in growth inhibition and cancer cell differentiation makes it a potential target for therapeutic interventions.

Research and Applications

Human recombinant PA2G4 is used in research to study its function and mechanism in cellular processes. It is also utilized in drug development and cancer research due to its involvement in growth regulation and signal transduction pathways.

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.