PAX8 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Characteristics

PAX8 recombinant monoclonal antibodies are produced via genetic engineering, involving the cloning of antibody DNA sequences into expression systems. Key features include:

ParameterDetails
Target ProteinPAX8 (62 kDa nuclear transcription factor; UniProt: Q06710)
ImmunogenRecombinant human PAX8 fragment (aa 60–261)
Host/IsotypeMouse (IgG2b κ) or rabbit (IgG)
LocalizationNuclear staining in PAX8-positive cells
ApplicationsImmunohistochemistry (IHC), Western blot, therapeutic target studies

Key Variants

  • Mouse Monoclonal (e.g., PAX8/1491): Recognizes 62 kDa protein; validated for renal and thyroid tumor detection .

  • Rabbit Recombinant (e.g., PAX8/2774R): Biotinylated or fluorescently labeled; cross-reactive with canine tissues .

Tumor Diagnosis

PAX8 antibodies are pivotal in distinguishing primary vs. metastatic tumors:

Tumor TypePAX8 PositivityClinical Utility
Thyroid follicular neoplasms98.6–100% Confirming thyroid origin
Ovarian serous/endometrioid carcinomasUp to 100% Differentiating from mucinous or metastatic tumors
Renal cell carcinomas82.6–97.8% Identifying renal origin
Urothelial neoplasms2.3–23.7% Limited utility due to low expression

Key Findings

  • High Specificity: Monoclonal antibodies (e.g., PAX8/1491) show minimal cross-reactivity with PAX5/PAX6, unlike polyclonal variants .

  • Negative Markers: Absent in breast, prostate, pancreatic, and pulmonary adenocarcinomas .

Mechanistic Studies

  • Epithelial-Mesenchymal Transition (EMT): PAX8 is implicated in promoting EMT in cancers, though its role remains under investigation .

  • Angiogenesis: Potential involvement in tumor vascularization is being explored .

Orthogonal Validation

PAX8 antibody specificity is confirmed via:

  • RNA-Seq Correlation: Concordance with Human Protein Atlas (HPA) and GTEx datasets .

  • Independent Antibodies: Cross-verification with alternative clones to exclude cross-reactivity .

Validation MethodOutcome
RNA ScreeningPAX8 mRNA detected only in tissues with confirmed antibody staining
Antibody ComparisonNuclear staining validated; cytoplasmic signals deemed non-specific

Limitations

  • Polyclonal Antibodies: Cross-reactivity with PAX5/PAX6 in lung/thymic tumors .

  • Cytoplasmic Artifacts: Observed in inflammatory cells and gastric glands, requiring careful interpretation .

Antibody Formulations

ParameterMouse (PAX8/1491) Rabbit (PAX8/2774R)
Concentration0.2 mg/mlVaries (check vial label)
Preservative0.05% Sodium AzideAzide-free options available
Storage2–8°C (with azide)-20°C to -80°C (without azide)
IHC Dilution1–2 µg/ml1:50–1:200

Tumor Expression Patterns

A 2024 study analyzing >17,000 tumors revealed:

Tumor CategoryStrong PAX8 PositivityWeak/Moderate Positivity
Thyroid follicular tumors15/16 (93.8%)1/16 (6.3%)
Gynecological carcinomas32/32 (100%)0/32
Renal cell carcinomas123/125 (98.4%)2/125 (1.6%)
Urothelial tumors5/21 (23.8%)18/21 (85.7%)

Data adapted from Gorbokon et al. (2024) .

Therapeutic Potential

PAX8 is under investigation as a target for:

  • Oncolytic Viruses: Engineered to lyse PAX8-expressing cells .

  • Targeted Therapies: Antibody-drug conjugates (ADCs) or CAR-T cell therapies .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The PAX8 Recombinant Monoclonal Antibody is produced through in vitro expression systems. This involves cloning PAX8 antibody DNA sequences from immunoreactive rabbits. The immunogen used is a synthesized peptide derived from the human PAX8 protein. Subsequently, the genes encoding the PAX8 antibodies are incorporated into plasmid vectors, which are then transfected into host cells for antibody expression. The PAX8 Recombinant Monoclonal Antibody is then purified via affinity chromatography and undergoes extensive testing for functionality in ELISA and IHC applications. It exhibits reactivity with the human PAX8 protein during these assessments.

PAX8 is a transcription factor that plays a crucial role in the development and function of various tissues and organs, including the thyroid gland, kidneys, and female reproductive system. It may also influence other tissues. Its regulatory function in gene expression is vital for tissue differentiation, development, and proper function.

Form
Liquid
Lead Time
Generally, we are able to dispatch products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery time details.
Synonyms
Paired box protein Pax-8, PAX8
Target Names
Uniprot No.

Target Background

Function
PAX8 serves as a transcription factor, specifically regulating the expression of genes exclusively found in thyroid cells. This ensures the maintenance of functional differentiation within these cells.
Gene References Into Functions
  1. PAX8-PPARG fusions may not play a significant role in the tumorigenesis of pediatric follicular thyroid carcinoma. PMID: 28621837
  2. Several novel PAX8 mutations have been identified in congenital hypothyroidism patients. These mutations impair the binding or activating capabilities of PAX8 at the promoters of target genes, such as thyroglobulin and thyroperoxidase. PMID: 28060725
  3. Data suggests that PAX8-regulated genes and pathways could serve as additional targets for the treatment of ovarian carcinoma. PMID: 27259239
  4. PAX8 exhibits a cell-specific role in governing proliferation and migration in nontransformed ovarian surface epithelial cells compared to oviductal cells. However, its reduction in serous cancer cell lines suggests a common mechanism for decreasing cell survival. PMID: 27129161
  5. Findings demonstrate that iodinated TG in thyroid follicular lumen regulates TTF-1 and PAX8 expression through thyroid stimulating hormone/thyroid stimulating hormone receptor (TSH/TSHR) mediated cAMP-PKA and PLC-PKC signaling pathways. PMID: 28322461
  6. Case Report: Primary seminal vesicle carcinoma shows strong and diffuse nuclear labeling for PAX8. PMID: 28506732
  7. PAX8 is expressed in both benign and malignant mesothelium. Loss of BAP1 is highly specific for malignant peritoneal neoplasms, aiding in differentiating between benign mesothelial proliferations and ovarian serous tumors. PMID: 28877056
  8. While the exact biological function remains to be investigated, findings suggest a possible association of PAX8eQTLs in lncRNA AC016683.6 with hepatocellular carcinoma prognosis in the Chinese population. PMID: 28339471
  9. Findings indicate significant PTC-associated dysregulation of several PAX8 target genes, supporting the notion that PAX8-regulated molecular cascades play crucial roles during thyroid tumorigenesis. PMID: 27249794
  10. Putative PAX8 target genes are enriched for common serous epithelial ovarian cancer risk variants. PMID: 28103614
  11. PAX8 immunostain is negative in most cervical cell carcinomas and is less frequently expressed in endocervical adenocarcinomas compared to the previously reported high sensitivity for ovarian and endometrial adenocarcinomas. PMID: 27362905
  12. The study postulated that when co-expressed, both TTF-1 and PAX-8 exhibit anti-proliferative and anti-tumorigenic properties up to a threshold expression level. Beyond that, they can induce pro-tumorigenic effects in thyroid carcinomas. PMID: 27573549
  13. Direct sequencing of the PAX8 gene revealed a novel single nucleotide substitution (c.162 A>T) in exon 2 that resulted in the substitution of the normal serine 54 with a cysteine (S54C). This mutation was associated with elevated serum TSH levels. PMID: 27207603
  14. This is the first report of PAX8 aberrant transcript production in cervical cancer. The reported PAX8 isoforms possess differential transactivation properties; therefore, besides being a helpful marker for cancer detection, PAX8 isoforms can plausibly exert differential regulation properties during carcinogenesis. PMID: 27175788
  15. PAX2, PAX8, and CDX2 immunostains were performed on the TMA slides. PMID: 26797858
  16. Pax8 gene rearrangement is associated with breast cancer. PMID: 27797226
  17. PAX8 was negative in all cases of pulmonary neuroendocrine carcinoma (PNEC), while positive in 86.4% of thymic cases (TNEC). TTF-1 positivity showed high sensitivity but low specificity for PNEC. Adding PAX8 negativity significantly increased the specificity. PAX8 positivity alone exhibited nearly 100% specificity and 86.4% sensitivity for TNEC. PMID: 27761900
  18. The study suggested that PAX8 eQTLs SNPs (rs4848320 and rs1110839) located in lncRNA PAX8-AS1 might predict a decreased risk of cervical cancer. PMID: 27225188
  19. High levels of mRNA for both PAX8 are associated with benign thyroid lesions compared to malignant ones. PMID: 26370671
  20. Results indicate that the presence of PAX8 immunoreactivity in an undifferentiated brain tumor lacking gliofibrillary acidic protein expression should prompt consideration of a metastatic tumor. PMID: 26371431
  21. Rete ovarii were positive for PAX-8, weakly positive for SF-1, and negative for PAX-2 and GATA-3. PMID: 26352548
  22. The study revealed that the PAX8 mutation rate is very low in thyroid dysgenesis patients in China. PMID: 26617871
  23. A substantial minority of solitary fibrous tumors express nuclear PAX8 and PAX2. PMID: 26404914
  24. PAX8 staining is helpful in distinguishing between primary thyroid squamous cell carcinoma and invasion or metastasis from extrathyroidal squamous cell carcinoma. PMID: 26354716
  25. PAX8 is expressed in the majority of benign, premalignant, and malignant endocervical glandular lesions. PMID: 26910219
  26. The PAX8 mutation rate was investigated among congenital hypothyroidism patients. PMID: 26362610
  27. The novel interplay between PAX8 and Neuropilin-2 was explored. PMID: 26030152
  28. A heterozygous transition in exon 3 of the PAX8 gene is associated with thyroid hypoplasia. PMID: 25720050
  29. miR-146b-3p binds to the 3'-untranslated region of PAX8 and sodium/iodide symporter. miR-146b and PAX8 regulate each other and share common target genes. PMID: 26282166
  30. PAX8-PPARgamma rearrangement was examined in 24 follicular thyroid carcinoma samples from Japanese patients. The fusion gene was detected in only one out of 24 (4%) cases. PMID: 25708358
  31. PAX8 protein expression was associated with germinal layers in forebrain and hindbrain development. PAX8 expression is linked to a better prognosis in medulloblastomas. PMID: 25287489
  32. PAX2 and PAX8 are useful biomarkers in the differential diagnosis of ovarian serous and mucinous tumors. PMID: 24992169
  33. PAX8 is a valuable immunohistochemical marker for differentiating seminal vesicle from prostate gland epithelium in prostate needle biopsies. PMID: 25153494
  34. PAX6 and PAX8 positivity was observed in metastatic pancreatic neuroendocrine tumors to the liver. PMID: 25433656
  35. In this series, PAX8/PPARgamma rearrangement found in thyroid nodules had a 100% predictive value for differentiated thyroid cancer. PMID: 24798894
  36. Compared to RAS or PAX8/PPARG-positive TCs, BRAFV600E or RET/PTC-positive thyroid cancers were more frequently associated with stage III/IV disease and recurrence. PMID: 26258321
  37. PAX8 (mAb) served as a specific marker in differentiating primary and extragenital metastatic mucinous ovarian tumors. PMID: 25827135
  38. PAX8 is frequently expressed by ovarian surface epithelial cells, and endogenous levels of PAX8 expression are non-transforming. PMID: 26079312
  39. Case Report: A novel PAX8 mutation is responsible for a severe form of dominantly inherited congenital hypothyroidism. The mutation appears to be linked to abnormalities of the urogenital tract. PMID: 23647375
  40. PAX8 immunoexpression was observed in five cases of alveolar rhabdomyosarcomas and three cases of embryonal rhabdomyosarcomas. Approximately one-third of malignant rhabdoid tumors were PAX2-positive and PAX8-positive. PMID: 24897005
  41. PAX8 is expressed in the vast majority of uterine adenocarcinomas. The level of expression, based on combined extent and intensity, is highest in endometrial serous carcinoma and lowest in endocervical adenocarcinoma. PMID: 25083965
  42. PAX8 is useful in distinguishing thymic carcinoma from poorly differentiated lung carcinoma. PMID: 23958552
  43. The reliability of PAX8 in determining tumor type or primary site was reviewed in 135 current clinical pelvic or abdominal lesions. PMID: 24857336
  44. The 5'-flanking region of the Wnt4 gene is responsive to Pax8. Pax8 modulates the expression of Wnt4 in thyroid cells. PMID: 25270402
  45. Results indicate that PAX8 plays a crucial role in the tumorigenic phenotype of ovarian cancer cells, identifying PAX8 as a potential new target for the treatment of ovarian cancer. PMID: 24766781
  46. PAX8 is expressed in the carcinomatous components of nearly all uterine malignant mesodermal mixed tumors, with expression in sarcomatous and undifferentiated components being less common and less extensive. PMID: 24901404
  47. This study confirms that PAX-8 expression is a useful diagnostic marker for renal cell carcinoma. PMID: 25315900
  48. PAX8 is increased in the majority of glioblastomas and promotes cell survival. PMID: 24602166
  49. The R133W-PAX8 variant is associated with a spectrum of phenotypes ranging from congenital hypothyroidism with thyroid hypoplasia to mild subclinical hypothyroidism. PMID: 25146893
  50. Data suggests that PAX8 provides signals for growth and motility of non-small cell lung cancer cells and is essential for MET and RON expression. PMID: 24628993

Show More

Hide All

Database Links

HGNC: 8622

OMIM: 167415

KEGG: hsa:7849

STRING: 9606.ENSP00000395498

UniGene: Hs.469728

Involvement In Disease
Hypothyroidism, congenital, non-goitrous, 2 (CHNG2)
Subcellular Location
Nucleus.
Tissue Specificity
Expressed in the excretory system, thyroid gland and Wilms tumors.

Q&A

What is PAX8 and why is it significant in research?

PAX8 is a 62 kDa nuclear protein belonging to the paired box (PAX) family of transcription factors. It plays a crucial role in thyroid follicular cell development and regulates the expression of thyroid-specific genes. The significance of PAX8 in research stems from its involvement in the development and functioning of specific tissues, particularly thyroid, kidney, and female reproductive organs. Mutations in the PAX8 gene have been associated with thyroid dysgenesis, thyroid follicular carcinomas, and atypical thyroid adenomas. Additionally, its differential expression pattern across various normal and neoplastic tissues makes it a valuable marker for diagnostic pathology .

What are the key differences between polyclonal and monoclonal PAX8 antibodies?

Polyclonal and monoclonal PAX8 antibodies differ significantly in their specificity and research applications:

CharacteristicPolyclonal PAX8 AntibodyMonoclonal PAX8 Antibody
SourceMultiple B-cell clonesSingle B-cell clone
Epitope recognitionMultiple epitopesSingle epitope
SpecificityLower; shows cross-reactivity with PAX5 and PAX6 proteinsHigher; superior specificity for PAX8
False positivesMore common in non-thyroid tumorsRare in non-thyroid tissues
Recommended useLess preferred for diagnostic applicationsPreferred for specific PAX8 detection

Research has demonstrated that monoclonal PAX8 antibodies show significantly higher specificity for thyroid tumors (98.0% positive rate) with minimal cross-reactivity. In contrast, polyclonal antibodies may yield false-positive results due to cross-reactivity with other PAX family proteins, particularly in lung carcinomas and thymic tumors .

In which tissues and tumor types is PAX8 expression most commonly observed?

PAX8 expression demonstrates a distinctive pattern across various tissues and tumors:

Tissue/Tumor TypePAX8 Expression RateIntensity
Thyroid neoplasms (follicular origin)98.6-100%Strong
Gynecological adenocarcinomasUp to 100%Variable
Renal tumors82.6-97.8%Variable
Urothelial neoplasms2.3-23.7%Variable
Ovarian serous carcinomasHigh percentageVariable
Ovarian endometrioid carcinomasHigh percentageVariable
Ovarian clear cell carcinomasHigh percentageVariable
Primary ovarian mucinous adenocarcinomasRarely positiveWeak

PAX8 is also expressed in non-neoplastic tissues including thyroid follicular cells, non-ciliated mucosal cells of the fallopian tubes, and renal tubules. Notably, it is absent in normal ovarian surface epithelial cells but present in simple ovarian inclusion cysts .

How should researchers optimize immunohistochemical protocols for PAX8 detection?

Optimizing immunohistochemical protocols for PAX8 detection requires careful consideration of several parameters:

  • Fixation and Tissue Processing:

    • Use 10% neutral buffered formalin for 24-48 hours.

    • Process tissues using standard paraffin embedding protocols.

    • Cut sections at 4-5 μm thickness for optimal staining.

  • Antigen Retrieval:

    • Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) is generally effective.

    • Pressure cooking for 15-20 minutes yields superior results compared to microwave methods.

  • Antibody Selection and Dilution:

    • Recombinant monoclonal rabbit antibodies (e.g., PAX8/2774R) generally provide more consistent results than mouse monoclonal antibodies.

    • Starting dilution recommendation: 1:100-1:200, with optimization based on specific tissue type.

    • Incubation time: 30-60 minutes at room temperature or overnight at 4°C.

  • Detection System:

    • Polymer-based detection systems provide better signal-to-noise ratio than avidin-biotin methods.

    • Hematoxylin counterstaining for 30-60 seconds provides optimal nuclear contrast.

  • Controls:

    • Positive control: Thyroid tissue or renal cell carcinoma (known PAX8 expressors).

    • Negative control: Omit primary antibody to assess background staining.

Assessment should focus on nuclear staining pattern, as PAX8 is a nuclear transcription factor. Any cytoplasmic staining should be considered non-specific .

What are the best practices for validating PAX8 antibody specificity in experimental contexts?

Validating PAX8 antibody specificity is essential to avoid misinterpretation of results. Researchers should implement the following validation approaches:

  • Multiple Antibody Comparison:

    • Test both monoclonal and polyclonal antibodies in parallel on the same tissues.

    • Compare PAX8 antibodies from different clones (e.g., PAX8/2774R vs. PAX8/1492).

    • Include related antibodies (PAX5, PAX6) to assess cross-reactivity.

  • Molecular Validation:

    • Implement in-situ hybridization for PAX8 mRNA (e.g., RNAscope technology) to confirm protein detection.

    • Positive staining for protein without mRNA detection suggests potential cross-reactivity.

  • Western Blot Verification:

    • Confirm antibody specificity by Western blot using known PAX8-expressing cell lines.

    • The expected molecular weight is approximately 48 kDa, although observed bands often appear at 60-62 kDa due to post-translational modifications.

  • Knockdown/Knockout Controls:

    • Test antibody on PAX8 knockdown or knockout cell lines to confirm specificity.

    • Absence of staining in knockout models confirms antibody specificity.

  • Tissue Panel Testing:

    • Use a diverse panel of tissues with known PAX8 status.

    • Include thyroid tissue (positive control) and tissues known to be PAX8-negative.

Studies have demonstrated that monoclonal PAX8 antibodies show superior specificity when validated using these approaches, while polyclonal antibodies may yield false positives due to cross-reactivity with PAX5 and PAX6 proteins, particularly in lung carcinomas and thymic tumors .

What are the appropriate applications for different PAX8 antibody conjugates?

Different PAX8 antibody conjugates offer distinct advantages for specific applications:

Conjugate TypeOptimal ApplicationsTechnical Considerations
UnconjugatedIHC-P, Western blotRequires secondary antibody; most versatile for routine diagnostics
Fluorescent dyes (CF® dyes)IF, Flow cytometryDirect detection; select wavelength based on microscopy setup
PerCP (Peridinin-chlorophyll)Flow cytometry, IFExcitation: 490 nm, Emission: 675 nm; good for multicolor analysis
Enzyme conjugates (HRP, AP)IHC, Western blotDirect visualization without secondary antibody
BiotinIHC, multiplex IHCAmplified signal; potential for higher background

Important considerations:

  • Blue fluorescent dyes (CF®405S, CF®405M) are not recommended for low-abundance targets due to lower fluorescence and higher non-specific background.

  • For multicolor flow cytometry, select conjugates with minimal spectral overlap with other fluorophores in the panel.

  • For multiplexed immunohistochemistry, consider using directly conjugated antibodies to avoid cross-reactivity issues with secondary antibodies.

  • For quantitative applications, enzyme conjugates with substrates producing soluble products are preferable .

How can PAX8 expression be utilized to differentiate between histologically similar tumors?

PAX8 immunostaining represents a powerful tool for distinguishing between histologically similar tumors, particularly in diagnostic pathology:

  • Renal Neoplasms vs. Adrenal Tumors:

    • Renal cell carcinomas show high PAX8 positivity (82.6-97.8%).

    • Adrenal cortical tumors are typically PAX8-negative.

    • This distinction is particularly valuable for retroperitoneal masses where anatomical origin is unclear.

  • Müllerian vs. Non-Müllerian Adenocarcinomas:

    • Ovarian, fallopian tube, and endometrial adenocarcinomas express PAX8.

    • Gastrointestinal and breast adenocarcinomas are typically PAX8-negative.

    • This differential expression pattern helps identify the primary site in metastatic adenocarcinomas.

  • Thyroid vs. Lung Neoplasms:

    • Thyroid tumors of follicular origin show near-universal PAX8 expression (98.6-100%).

    • Primary lung adenocarcinomas typically lack true PAX8 expression.

    • When using polyclonal antibodies, caution is needed as cross-reactivity with PAX5/PAX6 may cause false positives in lung tumors.

  • Urothelial Carcinoma vs. Prostate Carcinoma:

    • A subset of urothelial carcinomas (2.3-23.7%) express PAX8.

    • Prostate carcinomas are typically PAX8-negative.

    • This differential expression can help in distinguishing these entities in the genitourinary tract.

For optimal diagnostic accuracy, PAX8 should be used within a panel of immunohistochemical markers rather than in isolation, and monoclonal antibodies are strongly preferred due to their higher specificity .

What is the significance of PAX8 in understanding tumor histogenesis and molecular classification?

PAX8 expression patterns provide critical insights into tumor histogenesis and molecular classification:

  • Developmental Lineage Tracing:

    • PAX8 expression in tumors frequently recapitulates its expression in embryologic development.

    • The presence of PAX8 in ovarian epithelial tumors supports the theory that many "ovarian" carcinomas actually arise from fallopian tube epithelium or Müllerian inclusions rather than ovarian surface epithelium.

    • PAX8 positivity in nephroblastoma (Wilms tumor) reflects its role in renal development.

  • Molecular Subtypes and Classification:

    • In thyroid neoplasms, PAX8/PPARγ rearrangements define a subset of follicular carcinomas with distinct clinical behavior.

    • The pattern and intensity of PAX8 expression may correlate with molecular subtypes in renal and ovarian carcinomas.

    • PAX8 expression pattern can help differentiate histological subtypes, as seen in the high expression in serous, endometrioid, and clear cell ovarian carcinomas versus rare expression in mucinous subtypes.

  • Cell of Origin Determination:

    • PAX8 expression helps identify the cell of origin in poorly differentiated tumors.

    • For tumors of unknown primary, PAX8 positivity narrows the differential diagnosis to thyroid, renal, or Müllerian origin.

    • The absence of PAX8 in normal ovarian surface epithelium but its presence in inclusion cysts supports the dual origin theory of ovarian carcinomas.

  • Functional Implications:

    • As a transcription factor, PAX8 regulates specific downstream targets that may contribute to tumor phenotype.

    • PAX8-dependent pathways may represent potential therapeutic targets.

    • Loss or gain of PAX8 expression may contribute to dedifferentiation or transdifferentiation processes in tumor progression.

Understanding PAX8 expression in the context of tumor histogenesis provides not only diagnostic information but also insights into tumor biology that may guide therapeutic approaches .

How can researchers troubleshoot discrepancies between PAX8 protein detection and mRNA expression?

Discrepancies between PAX8 protein detection and mRNA expression represent a significant challenge that requires systematic troubleshooting:

  • Antibody Cross-reactivity Issues:

    • Studies have demonstrated that polyclonal PAX8 antibodies may cross-react with other PAX family proteins, particularly PAX5 and PAX6.

    • In one comprehensive study of thoracic tumors, no PAX8 mRNA expression was detected using RNAscope (in-situ hybridization) in tumors that tested positive with polyclonal PAX8 antibodies.

    • Approximately 31% of polyclonal PAX8 antibody-positive tumors (excluding thyroid tumors) showed positivity for PAX5 and/or PAX6, explaining the discrepancy.

  • Technical Validation Approach:

    • When discrepancies are observed, researchers should:
      a) Compare results using monoclonal versus polyclonal antibodies
      b) Perform parallel mRNA detection using in-situ hybridization
      c) Test the tissue with antibodies against other PAX family members
      d) Analyze PAX8 expression using RT-PCR or RNA sequencing

  • Interpretation Guidelines:

    • True PAX8 expression should show correlation between protein detection (preferably with monoclonal antibodies) and mRNA expression.

    • Positive protein staining without mRNA detection suggests potential cross-reactivity.

    • Weak mRNA with strong protein detection may indicate post-transcriptional regulation or protein stability differences.

  • Case Study Analysis:

    • In lung carcinomas and thymic tumors, discrepancies were resolved by demonstrating that positive staining with polyclonal PAX8 antibody was due to PAX5/PAX6 cross-reactivity.

    • Monoclonal PAX8 antibody correctly showed negative results in these tumors, consistent with mRNA findings.

This troubleshooting approach highlights the importance of using monoclonal PAX8 antibodies for accurate research and diagnostic purposes, particularly in tissues where PAX8 expression is not well-established .

What are the implications of PAX8 mutations and alterations for cancer development and progression?

PAX8 genetic alterations have significant implications for cancer development and progression:

  • PAX8-PPARγ Fusion:

    • The PAX8-PPARγ fusion gene results from a t(2;3)(q13;p25) chromosomal translocation.

    • Present in approximately 30-35% of follicular thyroid carcinomas and 2-13% of follicular adenomas.

    • The fusion protein acts as an oncogene through several mechanisms:
      a) Inhibition of wild-type PPARγ activity
      b) Activation of PAX8-responsive genes
      c) Promotion of cell growth and inhibition of apoptosis

  • PAX8 Gene Mutations:

    • Germline mutations in PAX8 are associated with congenital hypothyroidism and thyroid dysgenesis.

    • Somatic mutations have been identified in a subset of thyroid follicular carcinomas and atypical adenomas.

    • These mutations may lead to:
      a) Altered DNA binding capacity
      b) Changed protein-protein interactions
      c) Dysregulation of target gene expression

  • PAX8 Expression Alterations:

    • Overexpression of wild-type PAX8 is common in several cancer types:
      a) Thyroid carcinomas (especially well-differentiated types)
      b) Renal cell carcinomas
      c) Ovarian carcinomas

    • PAX8 upregulation may promote:
      a) Cell proliferation
      b) Invasion and migration
      c) Resistance to apoptosis
      d) Maintenance of cancer stem cell populations

  • Therapeutic Implications:

    • PAX8-dependent tumors may be vulnerable to:
      a) Direct PAX8 inhibition strategies
      b) Targeting PAX8-regulated pathways
      c) Immunotherapeutic approaches exploiting PAX8 as a tumor-associated antigen

    • PAX8 status may serve as a predictive biomarker for treatment response in certain cancer types.

Understanding the role of PAX8 alterations in cancer provides opportunities for developing targeted therapeutic strategies and improving prognostic assessment in PAX8-positive tumors .

What emerging technologies are enhancing the utility of PAX8 antibodies in cancer research?

Several cutting-edge technologies are expanding the applications of PAX8 antibodies in cancer research:

  • Multiplexed Immunofluorescence and Spectral Imaging:

    • Simultaneous detection of PAX8 with multiple markers in a single tissue section.

    • Allows assessment of PAX8 expression in specific cell populations identified by other markers.

    • Enables spatial relationship analysis between PAX8-positive cells and tumor microenvironment components.

    • Applications include tumor heterogeneity assessment and cell lineage tracing in complex tissues.

  • Mass Cytometry and Imaging Mass Cytometry:

    • Metal-tagged PAX8 antibodies enable highly multiplexed (30+ markers) analysis.

    • Single-cell resolution of PAX8 expression correlated with numerous other proteins.

    • Imaging mass cytometry provides spatial context while maintaining high multiplexing capacity.

    • Particularly valuable for analyzing rare cell populations within heterogeneous tumors.

  • Single-Cell Technologies:

    • Integration of PAX8 protein detection with single-cell RNA sequencing.

    • CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) allows correlation of PAX8 protein levels with whole-transcriptome analysis at single-cell resolution.

    • Reveals cell state transitions and differentiation trajectories in PAX8-positive cells.

  • Digital Pathology and AI-Based Analysis:

    • Automated quantification of PAX8 expression across large tissue areas.

    • Deep learning algorithms for pattern recognition of PAX8 staining in relation to morphological features.

    • Potential for computer-aided diagnosis based on PAX8 expression patterns.

    • Standardized analysis reduces inter-observer variability in PAX8 assessment.

These technologies are transforming PAX8 from a simple diagnostic marker to a powerful tool for understanding tumor biology, cellular heterogeneity, and disease progression at unprecedented resolution .

How can PAX8 expression analysis be integrated with other molecular markers for improved tumor classification?

Integration of PAX8 with other molecular markers creates powerful multiparametric approaches for tumor classification:

  • Integrated Diagnostic Algorithms:

    • Combining PAX8 with lineage-specific transcription factors:
      a) PAX8 + TTF-1: Distinguishes thyroid (both positive) from lung adenocarcinoma (TTF-1+/PAX8-)
      b) PAX8 + GATA3: Separates renal/Müllerian tumors (PAX8+/GATA3-) from urothelial/breast (PAX8-/GATA3+)
      c) PAX8 + WT1: Refines classification of serous carcinomas (both positive) vs. clear cell (PAX8+/WT1-)

    • Decision tree approaches incorporating PAX8 status significantly improve diagnostic accuracy.

  • Molecular Classification Systems:

    • PAX8 expression correlated with molecular subtypes defined by genomic analysis:
      a) In renal cell carcinoma: differential expression across TCGA molecular subtypes
      b) In ovarian carcinoma: correlation with TCGA genomic groups
      c) In thyroid carcinoma: association with RAS or BRAF mutation status

    • PAX8 status integrated with mutation profiles provides superior prognostic stratification.

  • Artificial Intelligence Applications:

    • Machine learning models incorporating:
      a) PAX8 immunohistochemistry
      b) Additional protein markers
      c) Morphological features
      d) Genomic data

    • These integrated models outperform single-marker approaches in challenging diagnostic scenarios.

  • Clinical Implementation Strategies:

    • Sequential testing algorithms starting with PAX8 and adding markers based on result

    • Standard immunohistochemistry panels tailored to specific differential diagnoses

    • Integration with molecular testing in reflex testing protocols

    • Standardized reporting systems incorporating multiple marker results

This integrated approach transforms PAX8 from a single biomarker to a component of comprehensive tumor classification systems with improved diagnostic precision and clinical relevance .

What are the methodological considerations for PAX8 antibody use in emerging single-cell analysis techniques?

Single-cell analysis techniques present unique challenges and opportunities for PAX8 antibody applications:

  • Antibody Validation for Single-Cell Applications:

    • Specificity requirements are even more stringent than for conventional applications.

    • Cross-reactivity can lead to significant misinterpretation of rare cell populations.

    • Validation approaches include:
      a) Testing on known positive and negative single-cell populations
      b) Correlation with mRNA expression at single-cell level
      c) Isotype and concentration-matched controls
      d) Comparison of multiple antibody clones

  • Optimizing Protocols for Mass Cytometry/CyTOF:

    • Metal conjugation may affect antibody binding characteristics.

    • Optimal metal selection to avoid signal spillover in detection channels.

    • Titration is essential to determine ideal concentration for single-cell resolution.

    • Cell fixation and permeabilization protocols must be optimized for nuclear transcription factors like PAX8.

    • Sample barcoding strategies to minimize batch effects.

  • Flow Cytometry Considerations:

    • Fluorophore selection based on instrument configuration and panel design.

    • Nuclear permeabilization protocols significantly impact PAX8 detection.
      a) Methanol-based methods (90% methanol) show superior results
      b) Commercial nuclear transcription factor buffers may require optimization

    • Doublet discrimination is crucial to avoid false positive signals.

    • Sequential gating strategies to identify true PAX8-positive populations.

  • Integration with Single-Cell Sequencing:

    • For CITE-seq applications:
      a) Antibody oligonucleotide tagging must not interfere with binding epitope
      b) Concentration optimization to avoid ADT (Antibody-Derived Tag) saturation
      c) Background correction methods to distinguish true signal from ambient noise

    • Computational analysis approaches for integrating protein and transcriptome data.

  • Sample Preparation Challenges:

    • Fresh tissue dissociation protocols must preserve PAX8 antigenicity.

    • Cryopreservation approaches that maintain nuclear antigen detection.

    • Balancing cell yield with epitope preservation during tissue disaggregation.

These methodological considerations are essential for generating reliable and interpretable data when applying PAX8 antibodies in cutting-edge single-cell analysis platforms .

What are the most significant unresolved questions regarding PAX8 biology and antibody applications?

Despite extensive research, several crucial questions about PAX8 and its antibodies remain unresolved:

  • PAX8 Function in Cancer Biology:

    • The precise role of PAX8 in promoting tumorigenesis outside of thyroid cancer remains poorly understood.

    • Whether PAX8 functions as an oncogene or tumor suppressor may depend on cellular context.

    • The downstream targets of PAX8 in different tissue types are incompletely characterized.

    • How PAX8 interacts with other transcription factors in regulatory networks requires further elucidation.

  • Technical Challenges in Antibody Development:

    • Creating monoclonal antibodies that recognize all PAX8 isoforms without cross-reactivity to other PAX family members remains difficult.

    • Standardization across antibody clones from different manufacturers is lacking.

    • Optimization for specific applications (e.g., ChIP-seq, proximity ligation assays) requires further development.

    • Quantitative assessment methods need refinement for more objective scoring systems.

  • Clinical Translation Gaps:

    • The prognostic significance of PAX8 expression levels (beyond binary positive/negative assessment) remains unclear.

    • Predictive value for treatment response in PAX8-positive tumors requires prospective validation.

    • The utility of circulating PAX8 protein or autoantibodies as liquid biopsy biomarkers is unexplored.

    • Implementation of standardized reporting systems across pathology laboratories has not been achieved.

  • Therapeutic Target Potential:

    • Whether PAX8 can be directly targeted therapeutically remains questionable.

    • The consequences of PAX8 inhibition in normal tissues expressing this transcription factor are unknown.

    • Identification of synthetic lethal interactions with PAX8 expression could provide alternative therapeutic strategies.

    • Immunotherapeutic approaches targeting PAX8 have not been thoroughly investigated.

Addressing these unresolved questions will require coordinated efforts combining basic science research with translational approaches and clinical validation studies .

How can researchers optimize experimental design for studies involving PAX8 expression in tumors?

Optimizing experimental design for PAX8 research requires careful consideration of several key factors:

  • Antibody Selection and Validation Strategy:

    • Use recombinant monoclonal antibodies with demonstrated specificity.

    • Include validation steps:
      a) Western blot confirmation of correct molecular weight
      b) Positive and negative tissue controls
      c) Correlation with mRNA expression
      d) Knockout/knockdown validation when possible

    • Compare results across multiple antibody clones when feasible.

    • Document clone, dilution, and detection system details in publications.

  • Comprehensive Sampling Approach:

    • Account for tumor heterogeneity through:
      a) Multiple sampling sites within tumors
      b) Inclusion of primary and metastatic sites when available
      c) Analysis of tumor margins and invasion fronts

    • Include matched normal tissues as controls.

    • Consider tissue microarray design with adequate cores per case (minimum 3).

    • Document sampling methodology clearly in research protocols.

  • Quantification and Scoring Systems:

    • Implement structured scoring approaches:
      a) Percentage of positive cells (not just positive/negative)
      b) Intensity scoring (0, 1+, 2+, 3+)
      c) H-score or Allred score calculation
      d) Digital image analysis when available

    • Include interobserver variability assessment for subjective scoring.

    • Document scoring methodology in detail in publications.

  • Multi-omics Integration:

    • Correlate protein expression with:
      a) mRNA expression (bulk or single-cell)
      b) Methylation status of PAX8 promoter
      c) Chromatin accessibility at PAX8 locus
      d) Mutation or copy number status

    • Implement pathway analysis to understand functional context.

    • Consider spatial transcriptomics to map expression patterns.

  • Clinical Correlation Design:

    • Collect comprehensive clinicopathological data:
      a) Detailed histomorphological assessment
      b) Treatment history
      c) Outcome data with adequate follow-up
      d) Response to specific therapies

    • Implement appropriate statistical methods for biomarker analysis.

    • Consider propensity score matching for retrospective studies.

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.