PABPN1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Development

PABPN1 recombinant monoclonal antibodies are produced by cloning antibody genes into mammalian expression systems (e.g., HEK293 cells) to express homogeneous antibodies targeting specific epitopes of PABPN1 . Unlike polyclonal antibodies, they minimize batch variability and enhance specificity. Key development steps include:

  • Immunogen Design: Synthetic peptides or fusion proteins derived from human PABPN1 (e.g., residues 4–51 or full-length protein) .

  • Host Systems: Primarily rabbit or mouse IgG isotypes .

  • Epitope Specificity: Targets include wild-type PABPN1 and mutant variants linked to oculopharyngeal muscular dystrophy (OPMD) .

Disease Mechanism Studies

  • OPMD Pathology: Antibodies like JM11-28 enable detection of nuclear aggregates formed by alanine-expanded PABPN1 in skeletal muscles, a hallmark of OPMD .

  • Gene Therapy Validation: Used in preclinical studies to verify knockdown of mutant PABPN1 and expression of codon-optimized replacements in mouse models .

Molecular Interactions

  • PAXT Complex Analysis: EP3000Y identifies PABPN1 as part of the poly(A) tail exosome targeting complex, linking RNA degradation to muscle atrophy .

  • Subcellular Localization: ICC/IF applications reveal PABPN1’s nuclear speckle localization and mislocalization in disease states .

Table 2: Key Studies Using PABPN1 Recombinant Monoclonal Antibodies

Study FocusAntibody UsedKey OutcomeReference
OPMD Gene TherapyJM11-28Confirmed AAV-mediated PABPN1 replacement normalizes muscle transcriptomes
Polyalanine Expansionα-Alanine AntibodyDetected A14–A17 PABPN1 in patient-derived myoblasts
RNA Surveillance MechanismsEP3000YIdentified PABPN1’s role in nuclear RNA quality control

Critical Insights:

  • Therapeutic Targeting: Dual gene therapy (knockdown + replacement) reduced insoluble aggregates by 85% in murine OPMD models .

  • Diagnostic Utility: Antibodies distinguishing wild-type vs. expanded PABPN1 (e.g., α-Alanine) improve OPMD biomarker detection .

Challenges and Future Directions

  • Specificity Limitations: Some clones cross-react with polyalanine stretches in unrelated proteins (e.g., RUNX2) .

  • Therapeutic Development: Recombinant antibodies are being explored as chaperones to dissolve PABPN1 aggregates in OPMD .

Product Specs

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

The PABPN1 Recombinant Monoclonal Antibody targets poly(A)-binding protein nuclear 1 (PABPN1), a multifaceted regulator of mRNA processing implicated in muscle wasting and atrophy. PABPN1 activates poly(A) polymerase, influencing poly(A) tail length on RNA transcripts and thereby impacting mRNA levels and stability through modulation of alternative polyadenylation site usage. Furthermore, it plays a crucial role in the processing of long non-coding RNA and short nucleolar RNA, as well as in nuclear surveillance mechanisms leading to RNA hyperadenylation and degradation. Physiologically, PABPN1 levels decline in humans from midlife onward, particularly within skeletal muscle. Pathologically, an expansion mutation in the PABPN1 gene is causative of oculopharyngeal muscular dystrophy (OPMD), a late-onset myopathy characterized by muscle wasting and atrophy.

This recombinant PABPN1 antibody was generated through a process involving PABPN1 gene acquisition, cloning into a suitable expression vector, transfection into mammalian cells, and subsequent purification of the functional antibody. Purification utilized a synthesized peptide derived from human PABPN1. The antibody demonstrates reactivity with human PABPN1 and is suitable for applications including ELISA, Western blotting (WB), immunohistochemistry (IHC), immunofluorescence (IF), and flow cytometry (FC).

Form
Liquid
Lead Time
Orders typically ship within 1–3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please consult your local distributor for precise delivery estimates.
Synonyms
Polyadenylate-binding protein 2 (PABP-2) (Poly(A)-binding protein 2) (Nuclear poly(A)-binding protein 1) (Poly(A)-binding protein II) (PABII) (Polyadenylate-binding nuclear protein 1), PABPN1, PAB2 PABP2
Target Names
PABPN1
Uniprot No.

Target Background

Function

PABPN1 plays a critical role in the 3'-end formation of mRNA precursors (pre-mRNA) by catalyzing the addition of a poly(A) tail (approximately 200–250 nucleotides) to the upstream cleavage product. It stimulates poly(A) polymerase (PAPOLA), enhancing the processivity of poly(A) tail elongation and regulating tail length. PABPN1 increases the RNA affinity of poly(A) polymerase. Its involvement extends to various stages of mRNA metabolism, including nucleocytoplasmic transport and nonsense-mediated decay (NMD) of mRNA. Synergistically with SKIP, PABPN1 activates E-box-mediated transcription via MYOD1, potentially regulating muscle-specific gene expression. It exhibits high-affinity binding to both poly(A) and poly(G) sequences, possibly protecting the poly(A) tail from degradation. Moreover, PABPN1 is a subunit of the trimeric poly(A) tail exosome targeting (PAXT) complex, which directs specific long, polyadenylated RNAs to the exosome for degradation. This exosome complex is essential for nuclear RNA degradation in eukaryotes, with substrate targeting facilitated by the cofactor MTREX, which links to RNA-binding protein adapters.

Gene References Into Functions
  1. Ala-expanded PABPN1 expression leads to the formation of nuclear aggregates preceding muscle weakness in OPMD. PMID: 27854203
  2. Characterization of expanded PABPN1 alleles in a Mexican OPMD cohort. PMID: 27980005
  3. CircPABPN1 binding to HuR prevents HuR binding to PABPN1 mRNA, reducing PABPN1 translation—a novel example of circRNA/mRNA competition for an RBP impacting translation. PMID: 28080204
  4. In vitro, PABPN1 strongly increases cognate poly(A) polymerase activity, unlike Pab2's effect on Pla1. PMID: 28096519
  5. The mRNA degradation PAXT complex comprises ZFC3H1 as a central link between MTR4 and nuclear PABPN1. PMID: 27871484
  6. PABPN1 aggregates trap TNNT3 pre-mRNA, displacing it from nuclear speckles and altering SC35-mediated splicing. PMID: 27507886
  7. Cytokine levels are unaltered in pre-symptomatic expPABPN1 carriers, suggesting potential as biomarkers for OPMD muscle weakness. Correlation patterns of expression levels may serve as novel disease state indicators. PMID: 27506982
  8. A large cohort study showed correlations between age at diagnosis, symptom severity, and PABPN1 repeat number in heterozygous and homozygous OPMD patients, with homozygous patients exhibiting a more severe phenotype, indicating a gene-dose effect. PMID: 28011929
  9. A polyadenylation-dependent 3' end maturation pathway for human telomerase RNA relies on PABPN1 and PARN. PMID: 26628368
  10. Validation of ribozymes targeting a mutant PABPN1 transcript. PMID: 26527730
  11. Investigation into the role of PABPN1 RNP domain stability and domain swapping in fibril formation. PMID: 26267866
  12. The PABPN1 and PAP-mediated RNA decay (PPD) pathway involves PABPN1, poly(A) polymerase (PAP), and the nuclear exosome complex. PMID: 26484760
  13. PABPN1 rescues several cytopathological features of TDP-43 proteinopathy by enhancing turnover of pathogenic proteins. PMID: 26130692
  14. PABPN1 inhibits expression of transcripts with proximal polyadenylation signals, possibly related to its role in RNA degradation. PMID: 25906188
  15. Phosphorylation analysis of PALB2 reveals roles of Ser-157 and Ser-376 in cellular responses to genotoxic stress. PMID: 26420486
  16. Impaired mRNA cleavage is an early defect in OPMD due to alanine-expanded PABPN1. PMID: 25816335
  17. Intron 6 of the PABPN1 gene is required for autoregulation. PMID: 25963658
  18. PABPN1's ability to promote splicing requires its RNA-binding and, to a lesser extent, PAP-stimulatory functions. PMID: 25896913
  19. ARIH2 E3 ligase regulates PABPN1 protein accumulation and aggregation. PMID: 24486325
  20. Loss of PABPN1, a suppressor of alternative polyadenylation, may promote tumor aggressiveness by releasing cancer cells from microRNA-mediated gene regulation. PMID: 24975429
  21. PABPN1 levels regulate muscle cell aging; OPMD represents accelerated muscle aging. PMID: 23793615
  22. While PABP4 nuclear translocation and increased PABP5 cytoplasmic abundance may compensate for PABPN1 loss, they are insufficient to prevent apoptosis, suggesting an anti-apoptotic role for PABPN1. PMID: 23300856
  23. No correlation found between muscle weakness, PABPN1 repeat frequency, and nuclear inclusion frequency/size. PMID: 22231868
  24. PABPN1 promotes lncRNA turnover via a polyadenylation-dependent mechanism. PMID: 23166521
  25. Nuclear speckles are biogenesis sites of PABPN1 inclusions in OPMD. PMID: 22249111
  26. Alanine-expanded PABPN1 initiates p53-dependent apoptosis. PMID: 22519734
  27. Possible cognitive and psychological impairments in heterozygous OPMD patients. PMID: 21956377
  28. Fibrillar PABPN1 differs structurally from native PABPN1; the C-terminal domain is implicated in fibril formation. PMID: 22570486
  29. PABPN1 acts as a suppressor of alternative cleavage and polyadenylation. PMID: 22502866
  30. In myotubes, the soluble/insoluble ratio of expPABPN1 is significantly lower than that of WT PABPN1. PMID: 21854744
  31. Transportin binding may delay PABPN1 methylation until after nuclear import. PMID: 21808065
  32. A (GCG)11 expansion in the PABPN1 gene caused OPMD in a Chinese family. PMID: 21647273
  33. The autosomal dominant form of OPMD is caused by (GCG) repeat expansions in the PABPN1 gene. PMID: 11689481
  34. hnRNP A1 and A/B co-localize with mutant PABPN1 in insoluble intranuclear aggregates in COS-7 cells. PMID: 12945950
  35. Families with the (GCG)11(GCA)3(GCG) mutation share a common ancestral haplotype; mutation age estimated at 37–53 generations. PMID: 15694141
  36. OPMD case with a (GCG)9 expansion in one allele and a normal (GCG)6 allele. PMID: 15725589
  37. Expanded PABPN1, via its polyalanine tract, leads to inclusion formation and neurodegeneration. PMID: 15755680
  38. Cytoplasmic targeting of mutant PABPN1 suppresses aggregation and toxicity in OPMD. PMID: 16101680
  39. Nuclear recruitment of HSP70 and HSC70 reduces mutant PABPN1 aggregation. PMID: 16239242
  40. PABPN1 plays a role in myogenesis. PMID: 16378590
  41. Overexpression of wild-type or mutant PABPN1 slows cell proliferation; this, along with apoptosis, may contribute to the late onset of OPMD. PMID: 16860991
  42. OPMD mutation provokes premature senescence in myoblasts, possibly due to intranuclear aggregates. PMID: 17005403
  43. The N-terminal domain of PABPN1 plays a role in OPMD. PMID: 17229142
  44. Nuclear aggregation in OPMD may represent an active process of sequestering and inactivating soluble toxic expPABPN1. PMID: 17418585
  45. OPMD case report in Italy. PMID: 18175083
  46. Wild-type PABPN1 overexpression reduces mutant PABPN1 toxicity in OPMD models. PMID: 18178579
  47. Unique PABPN1 gene mutation in a Bulgarian OPMD family. PMID: 18274805
  48. Induction of HSP27, HSP40, and HSP105 in mutant PABPN1-expressing cells. PMID: 18343218
  49. PABPN1 transgenic nematodes exhibit muscle cell degeneration and abnormal motility. PMID: 18397876
Database Links

HGNC: 8565

OMIM: 164300

KEGG: hsa:8106

STRING: 9606.ENSP00000216727

UniGene: Hs.707712

Involvement In Disease
Oculopharyngeal muscular dystrophy (OPMD)
Subcellular Location
Nucleus. Cytoplasm. Nucleus speckle.
Tissue Specificity
Ubiquitous.

Q&A

What is the difference between polyclonal and monoclonal antibodies for PABPN1 detection?

Polyclonal antibodies against PABPN1, like the α-alanine antibody described in recent literature, recognize multiple epitopes and are generated by immunizing animals (typically rabbits) with a synthetic antigen, such as a GST-tagged alanine peptide . These antibodies can detect polyalanine stretches in expanded PABPN1 (A14-A17) and other polyalanine proteins like RUNX2 .

In contrast, monoclonal antibodies for PABPN1 detection are derived from a single B-cell clone, ensuring recognition of a single epitope with high specificity and consistency between batches. For PABPN1 research, monoclonal antibodies offer advantages in experiments requiring precise quantification and consistent detection of specific forms of the protein.

When selecting between these antibody types, researchers should consider:

  • The specific experimental application (western blot, immunohistochemistry, flow cytometry)

  • Whether detection of wild-type vs. mutant PABPN1 is required

  • The need for batch-to-batch consistency in long-term studies

How do researchers validate the specificity of PABPN1 antibodies?

Validation of PABPN1 antibodies typically follows a multi-step approach:

  • Expression construct testing: Transfect cells (e.g., HEK293) with plasmids encoding PABPN1 with varying alanine repeat lengths (A10-wild type through A17) and perform immunoblotting to confirm specific detection of expanded forms .

  • Cross-reactivity assessment: Test against other polyalanine proteins (e.g., RUNX2 containing 17 alanine residues) to determine whether the antibody recognizes polyalanine stretches in multiple proteins or is specific to PABPN1 .

  • Tissue validation: Confirm detection capability in animal models of OPMD, including mouse and Drosophila models expressing alanine-expanded PABPN1 .

  • Knockout/knockdown controls: Verify antibody specificity using PABPN1 knockdown approaches, such as the shRNA3X system that targets endogenous PABPN1 .

  • Epitope mapping: Determine the exact binding site of the antibody, which is particularly important for distinguishing between wild-type and mutant PABPN1.

What are the key applications for PABPN1 antibodies in OPMD research?

PABPN1 antibodies enable several critical research applications:

  • Mutant protein detection: Antibodies that specifically recognize alanine-expanded PABPN1 allow for differentiation between wild-type and mutant forms, which has been challenging without tagged constructs .

  • Subcellular localization studies: Determine the localization patterns of wild-type versus mutant PABPN1, particularly in relation to nuclear aggregates and nuclear speckles .

  • Protein stability analysis: Investigate the half-life and steady-state levels of mutant PABPN1 compared to wild-type protein .

  • Therapeutic efficacy assessment: Evaluate the effectiveness of gene therapy approaches that aim to reduce mutant PABPN1 expression while maintaining functional protein levels .

  • Biomarker development: Monitor disease progression and treatment response in animal models and potentially in patient samples with expansions detectable by the antibody (A14-A18) .

How can researchers optimize immunodetection protocols for distinguishing between wild-type and alanine-expanded PABPN1?

Optimizing immunodetection protocols requires careful consideration of several factors:

  • Antibody selection: For western blotting, antibodies that recognize the polyalanine tract (like the α-alanine antibody) can detect expansions of A14 or larger, while failing to detect wild-type (A10) PABPN1 . This differential detection capability can be exploited for selective analysis.

  • Sample preparation: Nuclear fractionation may enhance detection sensitivity, as PABPN1 is predominantly nuclear and concentrating the fraction may improve signal-to-noise ratio.

  • Dual-color immunofluorescence: Combining antibodies that detect all PABPN1 forms with those specific to expanded forms allows visualization of the relative distribution and co-localization patterns.

  • Denaturing conditions: Modified SDS-PAGE conditions may be necessary to resolve the subtle size differences between wild-type and expanded PABPN1, which differ by only a few alanine residues.

  • Signal amplification techniques: For tissues with low expression levels, consider signal amplification methods such as tyramide signal amplification or proximity ligation assays.

When optimizing these protocols, researchers should systematically test variables including antibody concentration, incubation time and temperature, blocking agents, and detection methods to maximize specificity and sensitivity.

What are the considerations for using PABPN1 antibodies in gene therapy validation studies?

When using PABPN1 antibodies to validate gene therapy approaches, researchers should consider:

  • Knockdown verification: Antibodies that detect all PABPN1 forms are essential for confirming the efficacy of shRNA-mediated knockdown. In studies utilizing shRNA3X, a 35-45% decrease in endogenous PABPN1 expression was observed in treated muscles .

  • Replacement protein detection: When using a combined knockdown-replacement strategy, antibodies that distinguish between endogenous and exogenous PABPN1 are critical. For example, detection of MYC-tagged optPABPN1 confirmed expression of the replacement protein while endogenous PABPN1 was knocked down .

  • Temporal expression analysis: Serial sampling and antibody-based detection can track the kinetics of knockdown and replacement protein expression, informing optimal treatment schedules.

  • Off-target effects: Antibodies against potential off-target proteins should be employed to ensure specificity of the gene therapy approach.

  • Dose-response relationships: Antibody-based quantification at varying vector doses helps establish minimum effective dosage for therapeutic effect.

A comprehensive validation approach should include multiple detection methods, as demonstrated in studies where both western blotting and immunofluorescence were employed to verify PABPN1 knockdown and replacement protein expression .

How do PABPN1 antibodies contribute to understanding the molecular mechanisms of nuclear aggregate formation in OPMD?

PABPN1 antibodies have been instrumental in elucidating the composition and dynamics of nuclear aggregates in OPMD:

  • Aggregate composition analysis: Immunofluorescence using PABPN1-specific antibodies has revealed that both wild-type and mutant PABPN1 can be present in nuclear aggregates .

  • Co-localization studies: Combining PABPN1 antibodies with antibodies against other proteins helps identify additional components of aggregates, providing insights into pathological mechanisms.

  • Temporal aggregate formation: Time-course studies using PABPN1 antibodies can track the dynamics of aggregate formation, potentially revealing early intervention points.

  • Structure-function relationships: The α-alanine antibody specifically detecting expanded PABPN1 enables investigation of how alanine expansion affects protein-protein interactions and aggregate formation .

  • Disaggregation analysis: Antibody-based detection methods can assess the effectiveness of therapeutic approaches aimed at dissolving or preventing nuclear aggregates.

Research using the α-alanine antibody has demonstrated that it can distinguish alanine-expanded PABPN1 from the wild-type protein in muscle tissue from both fly and mouse models of OPMD, providing a valuable tool for studying mutant PABPN1 stability and localization independent of wild-type PABPN1 .

What techniques can be used to optimize western blot protocols for PABPN1 detection?

Optimizing western blot protocols for PABPN1 detection requires addressing several technical challenges:

ParameterStandard ConditionOptimized Condition for PABPN1
Gel percentage10-12%12-15% to better resolve small size differences
Sample preparationStandard lysisNuclear fraction enrichment
Transfer conditions100V, 1 hour30V overnight for improved transfer of aggregation-prone proteins
Blocking agent5% milk5% BSA to reduce background
Primary antibody dilution1:10001:500-1:2000 (antibody-dependent)
Incubation conditions1 hour, RTOvernight at 4°C for enhanced sensitivity
Detection methodStandard ECLEnhanced ECL or fluorescent detection for quantification

When detecting alanine-expanded PABPN1, researchers should note that the α-alanine antibody shows robust reactivity with A16- and A17-PABPN1, milder reactivity with A14- and A15-PABPN1, and no detection of wild-type (A10) or A13-PABPN1 . This differential detection should be considered when interpreting results from patient samples with varying expansion lengths.

For quantitative analysis, consider using fluorescent secondary antibodies and including standard curves with known quantities of recombinant PABPN1 protein.

What controls should be included when using PABPN1 antibodies in immunofluorescence studies?

A comprehensive set of controls is essential for reliable immunofluorescence results:

  • Positive controls:

    • Cells transfected with tagged PABPN1 constructs

    • Tissue from animal models known to express the target form of PABPN1

    • Recombinant PABPN1 protein spotted on slides

  • Negative controls:

    • PABPN1 knockdown cells (e.g., using shRNA3X)

    • Secondary antibody only

    • Isotype control antibody

    • Tissues from knockout models

  • Specificity controls:

    • Pre-absorption with immunizing peptide

    • Competitive binding with excess antigen

    • Parallel staining with multiple PABPN1 antibodies targeting different epitopes

  • Technical controls:

    • Nuclear counterstain to confirm subcellular localization

    • Co-staining with markers for nuclear speckles to assess physiological localization

    • Autofluorescence controls, especially important in muscle tissue

When studying nuclear aggregates, it's important to differentiate between physiological PABPN1 nuclear bodies and pathological inclusions by co-staining with markers such as SC35 (for nuclear speckles) and ubiquitin or HSP70 (for protein aggregates).

How can researchers quantitatively assess PABPN1 expression levels and aggregate formation?

Quantitative assessment of PABPN1 expression and aggregation requires rigorous methodological approaches:

  • Expression quantification:

    • Western blotting: Normalize PABPN1 signal to loading controls such as GAPDH or histone proteins for nuclear fractions. For gene therapy studies, compare endogenous PABPN1 levels across treatment groups, as demonstrated in muscles treated with shRNA3X showing 35-45% decrease in expression .

    • qRT-PCR: Measure mRNA levels using PABPN1-specific primers, with appropriate housekeeping gene normalization.

    • Flow cytometry: For cell culture models, antibody-based flow cytometry can provide population-level quantification.

  • Aggregate analysis:

    • Immunofluorescence quantification: Measure aggregate number, size, and intensity using automated image analysis software.

    • Aggregate isolation: Biochemical fractionation followed by western blotting to quantify aggregated versus soluble PABPN1.

    • Time-lapse imaging: For live-cell studies of aggregate dynamics using fluorescently tagged PABPN1 constructs.

  • In vivo assessment:

    • Tissue section analysis: Quantify percentage of nuclei containing aggregates across multiple sections.

    • Correlation with disease markers: Relate PABPN1 aggregation to muscle degeneration parameters or functional outcomes.

For gene therapy assessment, researchers have used dual detection methods: qRT-PCR to measure knockdown efficiency at the mRNA level and western blotting with tag-specific antibodies (MYC for optPABPN1, FLAG for expPABPN1) to confirm protein expression changes .

How might novel monoclonal antibodies advance the study of PABPN1 mutation-specific conformational changes?

Developing next-generation monoclonal antibodies could provide unprecedented insights into PABPN1 biology:

  • Conformation-specific antibodies: Antibodies that recognize specific conformational states of PABPN1 could distinguish between properly folded and misfolded variants, potentially identifying pre-aggregation intermediates.

  • Expansion-length specific antibodies: Creating a panel of monoclonal antibodies that differentially recognize varying alanine expansion lengths (A12, A13, A14, etc.) would allow more precise correlation between expansion size and disease phenotype.

  • Post-translational modification (PTM) antibodies: Antibodies targeting specific PTMs of PABPN1 could reveal how modifications regulate protein function and aggregation propensity.

  • Epitope binning approaches: Developing comprehensive epitope maps using multiple monoclonal antibodies could identify functional domains critical for PABPN1 activity and interactions.

  • Intrabodies for live-cell imaging: Engineered antibody fragments expressed intracellularly could track PABPN1 dynamics in real-time without the need for protein tagging.

Current α-alanine antibodies detect expansions of A14 or larger but cannot detect the A13 expansion found in the majority of OPMD patients . Development of antibodies with enhanced sensitivity to shorter expansions would significantly advance both research and diagnostic capabilities.

What role might PABPN1 antibodies play in validating emerging therapeutic approaches beyond gene therapy?

PABPN1 antibodies will be critical for evaluating multiple therapeutic strategies:

  • Small molecule screening: Antibody-based assays can assess compounds that prevent aggregation or enhance clearance of mutant PABPN1.

  • Protein quality control modulation: Antibodies detecting ubiquitinated or SUMOylated PABPN1 could evaluate therapies targeting protein degradation pathways.

  • RNA-based therapeutics: Similar to the validation of shRNA approaches, antibodies will be essential for confirming protein reduction with antisense oligonucleotides or CRISPR-based strategies.

  • Cell replacement therapies: Tracking PABPN1 expression in transplanted cells using specific antibodies will help assess therapeutic cell integration and function.

  • Combination therapies: Antibody-based detection methods will be crucial for understanding the interplay between multiple therapeutic approaches targeting different aspects of OPMD pathology.

In gene therapy validation studies, researchers have successfully used antibodies to confirm both knockdown of endogenous PABPN1 and expression of replacement proteins . Similar approaches will be essential for validating alternative therapeutic strategies, particularly those employing combined knockdown-replacement approaches.

How can PABPN1 antibodies contribute to developing biomarkers for OPMD progression and treatment response?

PABPN1 antibodies offer several avenues for biomarker development:

  • Circulating PABPN1 detection: Highly sensitive antibody-based assays could potentially detect PABPN1 released from damaged muscle tissue in blood or other accessible biofluids.

  • Muscle biopsy analysis: Standardized immunohistochemical protocols using PABPN1 antibodies could quantify nuclear aggregates as a measure of disease progression.

  • Single-cell analysis: Antibody-based flow cytometry or mass cytometry could assess PABPN1 expression heterogeneity within affected tissues.

  • Companion diagnostics: For gene therapy approaches, antibodies distinguishing between endogenous and replacement PABPN1 could serve as companion biomarkers for treatment monitoring.

  • Extracellular vesicle analysis: PABPN1 antibodies could detect protein in extracellular vesicles as potential accessible biomarkers.

While the α-alanine antibody's limited detection capability for shorter expansions (unable to detect A13-PABPN1) restricts its diagnostic utility in most OPMD patients , it remains valuable for research models and patients with larger expansions. Future development of antibodies with broader detection capabilities would enhance their biomarker potential.

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.