PAX8 Human

Paired Box 8 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

PAX8 contains a conserved 128-amino-acid paired domain for DNA binding and a partial homeodomain. Alternative splicing generates multiple isoforms, including:

IsoformFeaturesExpression
PAX8aFull-length (450 aa)Thyroid, kidney
PAX8bLacks 63-aa serine-rich C-terminal domainKidney, Wilms' tumors
PAX8cTruncated transactivation domainOvarian cancer

These isoforms exhibit tissue-specific functions, with PAX8a being dominant in thyroid development .

Physiological Roles

PAX8 is essential for:

  • Thyroid Development: Activates TG (thyroglobulin), TPO (thyroid peroxidase), and SLC5A5 (sodium/iodide symporter) .

  • Kidney Organogenesis: Cooperates with PAX2 to regulate nephron progenitor cells .

  • Reproductive System: Maintains Müllerian duct-derived tissues (e.g., fallopian tubes, endometrium) .

Mice lacking Pax8 exhibit thyroid agenesis and kidney defects, confirming its developmental necessity .

Clinical Significance in Congenital Hypothyroidism

Over 15 PAX8 mutations are linked to congenital hypothyroidism (incidence: 1:2,000–4,000 births). Key mutations include:

MutationFunctional ImpactClinical Phenotype
G41VDisrupts DNA bindingThyroid hypoplasia, severe hypothyroidism
D94NImpairs TPO activationMild hypothyroidism
R31HReduces coactivator recruitmentEuthyroid goiter

These mutations reduce thyroid hormone synthesis by 30–70% in vitro .

Oncogenic Mechanisms in Cancer

PAX8 is reactivated or mutated in multiple cancers:

Thyroid Cancer

  • Follicular Thyroid Carcinoma (FTC):

    • PAX8-PPARγ fusion (30–40% of FTCs) drives proliferation via PPARγ signaling dysregulation .

    • Mutually exclusive with RAS mutations, indicating distinct oncogenic pathways .

Clear Cell Renal Cell Carcinoma (ccRCC)

  • PAX8 cooperates with HIF2A to activate CCND1 (cyclin D1) and MYC, promoting cell cycle progression .

  • The protective allele rs7948643 reduces PAX8 binding at CCND1 enhancers, lowering ccRCC risk .

Ovarian Cancer

  • PAX8 sustains tumor growth via the PAX8-FGF18 autocrine loop .

  • HDAC inhibitors (e.g., panobinostat) suppress PAX8 by altering H3K27 acetylation at super-enhancers .

Therapeutic Implications

TargetStrategyEfficacy
PAX8-PPARγPPARγ agonists (e.g., pioglitazone)Preclinical tumor reduction
HDACsPanobinostat, romidepsin60–70% reduction in ovarian tumor growth
PAX8-DNA bindingCRISPR knockoutApoptosis induction in renal cancer

PAX8 immunohistochemistry is also a diagnostic marker for renal/ovarian cancers .

Research Frontiers

  • Gene-Environment Interactions: How PAX8 mutations synergize with iodine deficiency in thyroid dysgenesis .

  • Epigenetic Regulation: Role of PAX8 in maintaining super-enhancers during metastasis .

  • Therapeutic Resistance: PAX8-mediated survival pathways in chemotherapy-resistant tumors .

Product Specs

Introduction
PAX8, also referred to as paired box gene 8, is crucial for thyroid follicular cell development and the expression of thyroid-specific genes. This protein forms complexes with transcription factors TTF-1 and TTF-2 to activate thyroid follicular cell differentiation and thyroid hormone production. This regulation is achieved by controlling the expression of genes such as NIS (sodium iodide symporter), TPO (thyroid peroxidase), TG (thyroglobulin), and TSHR (thyrotropin receptor). Mutations in the PAX8 gene are linked to CHNG2, a disorder characterized by congenital hypothyroidism without goiter.
Description
Recombinant Human PAX8 protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 310 amino acids, with a sequence spanning from amino acid 1 to 287, and has a molecular weight of 33.4 kDa. The protein is engineered with a 23 amino acid His-tag fused to its N-terminus. Purification is carried out using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The PAX8 protein solution is provided at a concentration of 0.25 mg/ml and is formulated in a buffer consisting of Phosphate buffered saline (pH 7.4), 20% glycerol, and 1 mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended periods, storage at -20°C is recommended. To ensure long-term stability, adding a carrier protein like HSA or BSA (0.1%) is advisable. Repeated freezing and thawing should be avoided.
Purity
The purity of the protein is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
Paired Box 8, Paired Box Gene 8, PAX8.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMPHNSIR SGHGGLNQLG GAFVNGRPLP EVVRQRIVDL AHQGVRPCDI SRQLRVSHGC VSKILGRYYE TGSIRPGVIG GSKPKVATPK VVEKIGDYKR QNPTMFAWEI RDRLLAEGVC DNDTVPSVSS INRIIRTKVQ QPFNLPMDSC VATKSLSPGH TLIPSSAVTP PESPQSDSLG STYSINGLLG IAQPGSDKRK MDDSDQDSCR LSIDSQSSSS GPRKHLRTDA FSQHHLEPLE CPFERQHYPE AYASPSHTKG EQEVNTLAMP MATPPTPPTA RPGASPTPAC

Q&A

What is the molecular structure and genetic organization of human PAX8?

PAX8 belongs to an evolutionarily conserved family of nine nuclear transcription factors (PAX1-PAX9) that play crucial roles in lineage-dependent regulation during embryogenesis. In humans, PAX8 contains an open reading frame of 450 amino acids with a 128 amino acid paired domain at its amino-terminal end. The human and mouse Pax8 proteins show remarkable 97.8% conservation and identical paired domains, underscoring its evolutionary importance. Alternative splicing results in variant transcripts, including one that removes a 63 amino acid serine-rich region from the carboxy terminus, making the truncated protein more similar to murine Pax2 . These structural characteristics are essential to understand when designing experiments targeting specific domains or splice variants.

What are the normal expression patterns of PAX8 during human development?

PAX8 exhibits highly specific spatiotemporal expression patterns during embryogenesis. It is restrictedly expressed in developing brain, thyroid, kidney, and the Müllerian tract (from which fallopian tubes, uterus, cervix, and upper third of vagina originate). During kidney development specifically, PAX8 expression is detected in condensed mesenchyme, comma-shaped bodies, and S-shaped bodies, contrasting with PAX2 which is expressed primarily in earlier differentiation stages in the induced, condensing mesenchyme . In brain development, PAX8 protein expression is associated with germinal layers in both forebrain and hindbrain. This expression pattern changes dynamically, decreasing over time in the external granule cell layer while increasing in the internal granule cell layer . Understanding these developmental expression patterns is crucial for investigating PAX8's role in congenital disorders and developmental pathologies.

How should researchers approach PAX8 detection in experimental settings?

Researchers should employ multiple complementary techniques for robust PAX8 detection:

MethodApplicationTechnical Considerations
ImmunohistochemistryTissue expression analysisScore only nuclear staining; evaluate extent and intensity
Western blottingProtein expression quantificationConfirm antibody specificity with known positive controls
RNAse protection analysisTranscript variant detectionCan detect both full-length and truncated transcripts
In situ hybridizationSpatial expression in tissue sectionsCrucial for developmental studies
RT-PCRmRNA expression analysisDesign primers to distinguish splice variants

When performing immunohistochemistry, researchers must be aware that only nuclear staining should be scored as positive, and both extent and intensity should be evaluated for complete characterization .

What is the diagnostic utility of PAX8 immunostaining in tumor classification?

PAX8 serves as a highly specific and sensitive diagnostic marker for tumors originating from thyroid, renal, Müllerian, and thymic tissues. A comprehensive analysis of 1,357 tumors revealed the following PAX8 positivity rates:

Tumor TypePAX8 Positive CasesPercentage
Thyroid tumors60/6691%
Renal cell carcinomas158/17690%
Renal oncocytomas13/1681%
High-grade ovarian serous carcinomas164/16599%
Nonserous ovarian epithelial neoplasms32/4971%
Cervical epithelial lesions10/1191%
Endometrial adenocarcinomas152/15598%

Importantly, only 4% (30/719) of tumors from other origins showed any PAX8 positivity, making it an excellent marker for determining primary tumor site in metastatic disease . When designing diagnostic panels, researchers should consider combining PAX8 with other lineage-specific markers, such as thyroid transcription factor-1, RCC, and Wilms tumor-1, to achieve maximum diagnostic accuracy.

How does PAX8 function as a lineage-dependency factor in ovarian cancer?

PAX8 functions as a prototype lineage-survival oncogene in epithelial ovarian cancer (EOC), demonstrating functional essentiality regardless of distinct somatic alterations or histologies. Project Achilles data supports that PAX8 is frequently upregulated and functionally required in major subsets of ovarian cancer. Mechanistically, PAX8 regulates a network of genes (the PAX8 regulon) that mediates its oncogenic functions. GSEA analysis following PAX8 knockdown reveals suppression of cell cycle-related pathways and multiple signaling modules promoting tumor metastasis . Experimental approaches to study this lineage dependency include:

  • RNAi-mediated knockdown to assess effects on proliferation and metastatic potential

  • CRISPR-Cas9-mediated knockout using independent sgRNAs

  • Microarray or RNA-seq analysis following PAX8 modulation to identify downstream targets

  • Growth assays and crystal violet staining to quantify dependency effects

These methodological approaches are essential for researchers investigating PAX8 as a potential therapeutic target in gynecologic malignancies.

What is the prognostic significance of PAX8 expression in different tumor types?

PAX8 expression demonstrates context-dependent prognostic significance across different tumor types. In medulloblastomas, high PAX8 expression is associated with significantly better patient outcomes:

In vitro functional studies demonstrate that PAX8 knockdown increases proliferation and migration of medulloblastoma cell lines, suggesting that PAX8 suppresses tumorigenic properties in this context . This contrasts with its role in ovarian cancer, where it promotes tumorigenesis, highlighting the importance of conducting tissue-specific analyses when investigating PAX8 as a biomarker.

How can researchers effectively modulate PAX8 expression in experimental models?

Researchers can employ several complementary approaches to modulate PAX8 expression:

  • RNA interference: siRNAs targeting PAX8 have been successfully used in various cell lines, including OVTOKO, KURAMOCHI, HEY, and SKOV3, with phenotypic effects quantifiable by phase-contrast microscopy and crystal violet staining .

  • CRISPR-Cas9 gene editing: Complete PAX8 knockout can be achieved using two independent sgRNAs to minimize off-target effects. Western blotting should be performed to confirm knockout efficiency .

  • Inducible expression systems: For gain-of-function studies, doxycycline-inducible vectors carrying PAX8 cDNA can provide temporal control over expression.

  • Domain-specific mutations: To study specific PAX8 functions, mutations in the paired-box domain or other functional regions can be introduced using site-directed mutagenesis.

When designing these experiments, researchers should consider cell type-specific dependencies and include appropriate controls to account for potential compensation by other PAX family members.

What are the methodological approaches to identify the PAX8 regulon in tissue-specific contexts?

Identifying the PAX8 regulon (genes regulated by PAX8) requires a multi-omics approach:

  • Transcriptome analysis: Perform microarray or RNA-seq on cells with PAX8 knockdown or knockout compared to controls. Hierarchical clustering of differentially expressed genes can reveal distinct expression patterns .

  • Chromatin immunoprecipitation sequencing (ChIP-seq): Map direct PAX8 binding sites across the genome to distinguish between direct and indirect target genes.

  • Gene Set Enrichment Analysis (GSEA): Apply GSEA to identify significantly altered signaling modules and biological pathways following PAX8 modulation. This has revealed PAX8's role in regulating cell cycle and tumor metastasis-related pathways .

  • Integration with histone modification data: Since PAX8 interacts with epigenetic mechanisms, correlating PAX8 binding with histone marks can provide insights into its regulatory mechanisms.

  • Validation studies: Confirm key targets using reporter assays, site-directed mutagenesis of binding sites, and rescue experiments.

This comprehensive approach enables identification of tissue-specific PAX8 regulatory networks, essential for understanding its context-dependent functions.

How can researchers exploit PAX8's epigenetic vulnerabilities for therapeutic development?

PAX8-dependent tumors show vulnerability to epigenetic modulation, particularly through HDAC inhibitors. A high-throughput image-based screening approach with a library of 180 FDA-approved or clinically relevant compounds identified multiple HDAC inhibitors that significantly decreased PAX8/DAPI intensity ratios in cancer cells . Methodological considerations for investigating this vulnerability include:

  • High-content imaging: Quantify nuclear PAX8 staining normalized to DAPI across drug libraries.

  • Dose and time-dependent analyses: Western blotting following drug treatment (e.g., panobinostat) at various concentrations and timepoints can establish pharmacodynamic relationships .

  • Functional validation: Assess whether PAX8 downregulation mediates the anti-tumor effects of HDAC inhibitors through rescue experiments with PAX8 overexpression.

  • Combinatorial approaches: Test HDAC inhibitors in combination with other targeted therapies that may synergize with PAX8 inhibition.

  • Biomarker development: Identify predictive biomarkers of response to HDAC inhibition in PAX8-dependent tumors.

This research direction is particularly promising as multiple HDAC inhibitors have already earned FDA approval for other cancer types, potentially accelerating clinical translation.

What are the challenges and solutions in targeting transcription factors like PAX8?

Direct targeting of transcription factors presents several challenges:

ChallengePotential Solutions
Lack of enzymatic activityTarget protein-protein interactions using small molecules or peptide mimetics
Nuclear localizationDevelop nuclear-penetrating drug delivery systems
Redundancy with other PAX family membersDesign highly specific compounds that exploit unique structural features
Essential role in normal tissuesUse tumor-specific delivery systems or exploit context-dependent vulnerabilities
Structural complexityApply fragment-based drug discovery approaches

Alternative strategies include:

  • Targeting the PAX8 regulon: Identify and target critical downstream effectors that mediate PAX8's oncogenic functions.

  • Exploiting synthetic lethality: Identify genes that, when inhibited, cause selective lethality in PAX8-dependent cells.

  • Epigenetic modulation: As demonstrated with HDAC inhibitors, target epigenetic mechanisms that regulate PAX8 expression .

  • Degradation approaches: Develop PAX8-directed PROTACs (proteolysis targeting chimeras) to induce selective protein degradation.

How does PAX8 interact with developmental signaling pathways in normal and pathological contexts?

PAX8 demonstrates complex interactions with developmental pathways, particularly evident in medulloblastomas where PAX8 expression associates specifically with sonic hedgehog (SHH) and wingless int (WNT) subtypes but not with group 3 and 4 medulloblastomas . This subtype-specific expression pattern suggests co-regulation with or modulation of these developmental signaling cascades. Methodological approaches to investigate these interactions include:

  • Co-expression analyses in developmental and pathological tissues

  • Pathway inhibition studies to assess effects on PAX8 expression

  • ChIP-seq analyses to identify potential co-binding of PAX8 with SHH or WNT effectors

  • Genetic interaction studies using combined knockdown/knockout approaches

  • Developmental time-course analyses to map temporal relationships between pathway activation and PAX8 expression

Understanding these interactions may reveal how developmental programs are coopted during tumorigenesis and inform subtype-specific therapeutic approaches.

What are the most promising approaches to resolve contradictory roles of PAX8 across different tissues?

PAX8 demonstrates seemingly contradictory roles across different contexts—promoting tumorigenesis in ovarian cancer while suppressing it in medulloblastoma. Resolving these apparent contradictions requires:

  • Context-specific interactome mapping: Identify tissue-specific protein interaction partners through techniques like BioID or IP-MS.

  • Comparative epigenomic profiling: Analyze chromatin accessibility and histone modifications at PAX8 binding sites across different tissues.

  • Single-cell multi-omics approaches: Characterize PAX8 function at single-cell resolution to identify cell state-specific roles.

  • Developmental origin considerations: Investigate how cellular ontogeny influences PAX8 function, particularly regarding WNT and SHH pathway interactions.

  • Systematic domain function analysis: Assess whether different functional domains mediate context-specific activities.

These approaches can help resolve the molecular basis for PAX8's diverse functions and guide context-appropriate therapeutic strategies targeting PAX8 or its pathways.

Product Science Overview

Structure and Function

PAX8 typically contains:

  • Paired Box Domain: This domain is essential for DNA-binding activity. It consists of 128 amino acids and is folded into two β-sheets and six α-helices divided into two subdomains, PAI and RED. Mutations in these subdomains can disrupt DNA-binding activity and lead to congenital abnormalities .
  • Octapeptide: This is a conserved motif found in PAX proteins.
  • Paired-Type Homeodomain: This domain can serve as a second independent DNA-binding region .
Role in Development and Disease

PAX8 is crucial for the development of several organs, including the thyroid gland, kidneys, and reproductive system. It is involved in:

  • Thyroid Development: PAX8 is essential for the differentiation and function of thyroid follicular cells.
  • Kidney Cell Differentiation: It plays a role in the development of the kidneys.
  • Reproductive System Development: PAX8 is important for the development of the reproductive system, particularly the Müllerian ducts .
PAX8 in Cancer

PAX8 expression is deregulated in several types of cancer, including those arising from the reproductive tract. In cancer, PAX8 can:

  • Stimulate Proliferation: It promotes the growth of cancer cells.
  • Inhibit Apoptosis: PAX8 helps cancer cells evade programmed cell death.
  • Regulate Angiogenesis: It is involved in the formation of new blood vessels to supply the tumor .

PAX8 can interact with different protein partners during cancer progression and may exhibit significant function-altering alternative splicing. Moreover, the expression of PAX8 in cancer can serve as a biomarker for diagnostic and prognostic purposes .

Recombinant PAX8

Recombinant PAX8 is a form of the protein that is produced using recombinant DNA technology. It is typically expressed in Escherichia coli and purified for use in research. The recombinant protein often includes a His-tag for purification purposes and corresponds to specific amino acids of the human PAX8 protein .

Recombinant PAX8 is used in various research applications, including studies on gene regulation, protein-protein interactions, and the role of PAX8 in development and disease. It is also used to investigate the mechanisms of PAX8 function and its potential as a therapeutic target in cancer .

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.