PHYHD1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
The antibody is provided in phosphate buffered saline (PBS) with 0.02% sodium azide, 50% glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for specific delivery timelines.
Synonyms
MGC16638 antibody; OTTHUMP00000022302 antibody; OTTHUMP00000022303 antibody; OTTHUMP00000022304 antibody; OTTHUMP00000022305 antibody; OTTHUMP00000199642 antibody; PHYD1_HUMAN antibody; Phyhd1 antibody; Phytanoyl CoA dioxygenase domain containing 1 antibody; Phytanoyl CoA dioxygenase domain containing protein 1 antibody; Phytanoyl-CoA dioxygenase domain-containing protein 1 antibody
Target Names
PHYHD1
Uniprot No.

Target Background

Function

Isoform 1 exhibits alpha-ketoglutarate-dependent dioxygenase activity. It does not display detectable activity towards fatty acid CoA thioesters and is not expected to be active with phytanoyl CoA. Isoforms 2 and 3 likely lack enzymatic activity.

Gene References Into Functions
  1. PHYHD1A possesses the characteristic double-stranded beta-helix fold, along with Fe(II) and cosubstrate binding residues typical of 2-oxoglutarate dependent oxygenases. It catalyzes the conversion of 2-oxoglutarate to succinate and CO2 in an iron-dependent manner. PMID: 21530488
Database Links

HGNC: 23396

KEGG: hsa:254295

STRING: 9606.ENSP00000309515

UniGene: Hs.709447

Protein Families
PhyH family, PHYHD1 subfamily

Q&A

What is PHYHD1 and what are its key functional domains?

PHYHD1 (Phytanoyl-CoA Dioxygenase Domain Containing 1) is a 291 amino acid protein belonging to the PHYH family and PHYHD1 subfamily. It maps to human chromosome 9q34.11 and exists as three alternatively spliced isoforms. PHYHD1 likely functions as an alpha-ketoglutarate-dependent dioxygenase, participating in metal ion binding and oxidoreductase activity that acts on single donors with incorporation of two atoms of oxygen . It shows homology to PHYH (phytanoyl-CoA 2-hydroxylase), which catalyzes the initial alpha-oxidation step in phytenic acid degradation in peroxisomes. Research indicates PHYHD1 may also play a role in DNA methylation during early postnatal liver development and mammalian differentiation . The protein's functional domains include the phytanoyl-CoA dioxygenase domain, which is critical for its enzymatic activity.

What types of PHYHD1 antibodies are currently available for research applications?

Research laboratories have access to several types of PHYHD1 antibodies with varying characteristics:

Antibody TypeHost SpeciesClonalityTarget RegionApplicationsCross-Reactivity
C-Terminal antibodiesRabbitPolyclonalAA 231-260WB, EIAHuman
Full-length antibodiesMousePolyclonalAA 1-297WBHuman
Middle-region antibodiesRabbitPolyclonalAA 33-287WB, ELISA, IHCHuman
Monoclonal antibody (OTI1A6)Not specifiedMonoclonalNot specifiedNot specifiedHuman
Recombinant fusion protein antibodyRabbitPolyclonalFull proteinWB, IFHuman, Mouse, Rat

When selecting an antibody, researchers should consider target epitope, clonality, and validated applications to ensure experimental success. Each antibody has been validated for specific applications, though comprehensive validation across all potential methods may not be available for all products .

How does PHYHD1 antibody specificity differ between monoclonal and polyclonal options?

Monoclonal antibodies like PHYHD1 antibody (OTI1A6) recognize single epitopes with high specificity but potentially lower sensitivity . They provide consistent results between batches, making them valuable for long-term studies requiring reproducibility.

In contrast, polyclonal PHYHD1 antibodies, such as those targeting the C-terminal region (AA 231-260), recognize multiple epitopes, offering higher sensitivity but potentially more cross-reactivity . This makes polyclonal antibodies particularly useful for detecting low-abundance PHYHD1 protein but may require more rigorous validation.

For detecting alternatively spliced PHYHD1 isoforms, epitope selection becomes critical. Antibodies targeting conserved regions will detect all isoforms, while those recognizing variable regions can differentiate between specific isoforms. When researching PHYHD1 in Alzheimer's disease models, where upregulation has been observed, sensitivity considerations may influence antibody selection .

What are the optimal conditions for Western Blot detection of PHYHD1?

For successful Western Blot detection of PHYHD1 (calculated MW: 32kDa ), researchers should implement the following optimized protocol:

  • Sample preparation: Extract proteins using RIPA buffer supplemented with protease inhibitors to prevent degradation of PHYHD1, which has been shown to be susceptible to proteolytic cleavage.

  • Gel selection: Use 10-12% SDS-PAGE gels for optimal resolution of PHYHD1's 32kDa band.

  • Transfer conditions: Semi-dry transfer at 15V for 45 minutes or wet transfer at 100V for 1 hour using PVDF membrane (preferred over nitrocellulose for PHYHD1 detection).

  • Blocking: 5% non-fat dry milk in TBST for 1 hour at room temperature has shown superior results compared to BSA-based blocking for PHYHD1 antibodies.

  • Primary antibody incubation: Dilute antibody according to manufacturer recommendations (typically 1:500-1:2000) in blocking buffer and incubate overnight at 4°C. C-terminal antibodies (AA 231-260) have demonstrated consistent results in detecting full-length PHYHD1 .

  • Signal development: Both chemiluminescence and fluorescent secondary antibodies work well, with fluorescent detection offering better quantitative analysis for PHYHD1 expression level studies.

  • Controls: Include recombinant PHYHD1 protein as a positive control and tissues known to express PHYHD1 (human brain cortex samples are appropriate positive controls based on Alzheimer's research findings ).

Troubleshooting tip: If multiple bands appear, optimize antibody concentration and consider using gradient gels to better resolve PHYHD1 isoforms, which have been documented in research literature .

How should PHYHD1 antibodies be validated for cross-reactivity and specificity?

Comprehensive validation of PHYHD1 antibodies requires multiple approaches to ensure experimental reliability:

  • Knockout/knockdown validation: Compare staining patterns between wild-type samples and those with PHYHD1 gene knockout or siRNA knockdown. This is particularly important given PHYHD1's homology to other PHYH family members.

  • Peptide competition assay: Pre-incubate the antibody with the immunizing peptide (such as the C-terminal peptide for antibodies targeting AA 231-260 ) before application to samples. Signal disappearance confirms specificity.

  • Multiple antibody comparison: Use antibodies from different suppliers or those targeting different epitopes of PHYHD1 (e.g., C-terminal vs. full-length) and compare staining patterns.

  • Mass spectrometry verification: For critical research, perform immunoprecipitation with the PHYHD1 antibody followed by mass spectrometry analysis to confirm target identity.

  • Cross-species reactivity testing: While some PHYHD1 antibodies have been validated for human, mouse, and rat reactivity , testing on your specific samples remains essential due to potential species-specific isoform variations.

  • Recombinant protein controls: Use purified recombinant PHYHD1 protein as a positive control in Western blots to verify appropriate molecular weight detection.

This multi-faceted validation approach is particularly important for studies investigating PHYHD1's role in Alzheimer's disease, where specificity is crucial for accurately measuring expression changes in disease models .

What are the critical considerations for immunofluorescence studies using PHYHD1 antibodies?

When conducting immunofluorescence (IF) studies with PHYHD1 antibodies, researchers should address these methodology-specific considerations:

How can researchers interpret PHYHD1 expression changes in Alzheimer's disease models?

PHYHD1 has been identified as significantly upregulated in Alzheimer's disease (AD) frontal cortex, alongside other genes including C4A/C4B, CD74, and GFAP . When interpreting PHYHD1 expression changes in AD research:

  • Temporal analysis: Expression changes should be analyzed across disease progression stages. In App NL-G-F/NL-G-F mouse models, PHYHD1 upregulation correlates with Aβ accumulation beginning at 4-6 months of age .

  • Regional specificity: PHYHD1 upregulation shows regional variation, with microarray analyses confirming differential expression between temporal and frontal cortices. These patterns should be compared with Aβ deposition maps.

  • Network analysis: Interpret PHYHD1 changes within its molecular network context. PHYHD1 has been identified in inflammatory response networks alongside C4A/C4B, CD74, CTSS, CX3CR1, and others with direct or indirect connections to APP .

  • Correlation with pathological markers: When analyzing PHYHD1 expression:

Disease StagePHYHD1 ExpressionAssociated MarkersPathological Features
Early (Braak I-II)Beginning upregulationMinor CD74 increaseInitial Aβ deposition
Intermediate (Braak III-IV)Moderate upregulationIncreased GFAP, S100BWidespread Aβ plaques
Advanced (Braak V-VI)Significant upregulationHigh CD74, GFAP, C4A/C4BDense Aβ distribution, astrocyte activation

What are the methodological approaches for studying PHYHD1 protein-protein interactions?

To investigate PHYHD1's protein-protein interactions, particularly its reported interaction with Aβ42 relevant to Alzheimer's disease pathology , researchers should employ these complementary approaches:

  • Co-immunoprecipitation (Co-IP):

    • Use anti-PHYHD1 antibodies targeting different epitopes (C-terminal and full-length) to pull down protein complexes

    • Validate interactions bidirectionally with reverse Co-IP using antibodies against candidate interacting proteins

    • Include appropriate negative controls (IgG from the same species as the antibody)

    • For membrane-associated interactions, modify lysis conditions to include detergents that preserve membrane protein interactions

  • Proximity Ligation Assay (PLA):

    • Particularly useful for validating PHYHD1-Aβ42 interactions in situ in brain tissue

    • Requires two primary antibodies from different species (e.g., rabbit anti-PHYHD1 and mouse anti-Aβ42)

    • Quantify PLA signals in different brain regions to map interaction sites

  • Bioluminescence Resonance Energy Transfer (BRET):

    • Tag PHYHD1 with Renilla luciferase and potential partners with YFP

    • Measure energy transfer as evidence of direct protein interaction

    • Generate a BRET saturation curve to distinguish specific from non-specific interactions

  • Split-complementation assays:

    • Use BiFC (Bimolecular Fluorescence Complementation) with PHYHD1 fused to one half of a fluorescent protein

    • Particularly useful for visualizing subcellular localization of interactions

  • Mass spectrometry-based approaches:

    • BioID or APEX proximity labeling methods to identify proteins in close proximity to PHYHD1

    • Quantitative proteomics comparing PHYHD1 interactomes in normal versus AD models

  • Surface Plasmon Resonance (SPR):

    • Determine binding kinetics between purified PHYHD1 and potential partners

    • Particularly valuable for characterizing the PHYHD1-Aβ42 interaction reported in AD research

When investigating PHYHD1 interactions related to its alpha-ketoglutarate-dependent dioxygenase function , include cofactors (Fe(II), alpha-ketoglutarate) in binding assays to capture physiologically relevant interactions.

How can researchers address contradictory findings in PHYHD1 expression studies?

When reconciling inconsistent findings in PHYHD1 expression studies, researchers should implement these analytical approaches:

  • Antibody validation comparison:

    • Compare results obtained using different antibodies targeting distinct epitopes of PHYHD1

    • Verify whether antibodies detect all three alternatively spliced isoforms or are isoform-specific

    • Implement validation controls consistently across studies (knockout/knockdown controls)

  • Experimental condition harmonization:

    • Standardize sample preparation methods, particularly for metal ion-dependent proteins like PHYHD1

    • Document buffer compositions, especially regarding divalent cations and reducing agents

    • Consider enzymatic activity state (active vs. inactive) when interpreting localization differences

  • Cross-methodology verification:

    • Compare protein expression (Western blot/immunohistochemistry) with mRNA expression (qRT-PCR/RNA-seq)

    • For contradictory localization results, use multiple microscopy techniques (confocal, super-resolution)

    • Implement orthogonal approaches (e.g., CRISPR-tagged endogenous PHYHD1) to resolve antibody reliability issues

  • Context-dependent expression analysis:

    • Document cell type specificity when comparing brain tissue studies

    • Account for disease stage differences when comparing AD studies (early vs. late Braak stages)

    • Consider age-dependent changes in PHYHD1 expression profiles

  • Statistical and methodological reporting standards:

    • Implement comprehensive reporting of normalization controls

    • Document biological vs. technical replication clearly

    • Use appropriate statistical tests for the data distribution observed

For contradictory findings regarding PHYHD1's role in Alzheimer's disease , researchers should explicitly analyze how differences in patient cohorts (age, sex, comorbidities) and tissue sampling methods might influence results.

What are promising research directions for investigating PHYHD1's role in neurodegeneration?

Based on findings that PHYHD1 is upregulated in Alzheimer's disease cortex and directly interacts with Aβ42 , several promising research avenues emerge:

  • Mechanistic studies of PHYHD1-Aβ42 interaction:

    • Investigate whether PHYHD1's dioxygenase activity modifies Aβ42 structure or aggregation properties

    • Determine if this interaction is protective or pathological in neurodegeneration

    • Map the interaction domains using truncation mutants of both proteins

  • PHYHD1 knockout/knockdown in AD models:

    • Generate conditional PHYHD1 knockout in App NL-G-F/NL-G-F mouse models where PHYHD1 upregulation has been documented

    • Assess effects on Aβ accumulation, glial activation, and cognitive outcomes

    • Implement temporally controlled knockdown to distinguish developmental from disease-specific roles

  • Oxidative stress and PHYHD1 regulation:

    • Investigate how PHYHD1's oxidoreductase activity relates to oxidative stress in neurodegeneration

    • Examine potential crosstalk with the NFE2L2 pathway, which appears in the same network as PHYHD1 in AD contexts

    • Test whether PHYHD1 activity is modulated by age-related changes in iron homeostasis

  • Single-cell transcriptomics approaches:

    • Determine cell type-specific expression patterns of PHYHD1 in healthy and AD brains

    • Identify potential cell-autonomous versus non-cell-autonomous effects

    • Correlate with cellular stress response signatures

  • Translational biomarker development:

    • Evaluate PHYHD1 levels in CSF or plasma as potential biomarkers for AD progression

    • Correlate with existing AD biomarkers (Aβ42, tau, neurofilament light)

    • Develop PHYHD1 activity assays that might reflect disease state

These directions build upon the established connection between PHYHD1 and inflammatory networks in Alzheimer's disease , potentially providing new insights into disease mechanisms and therapeutic targets.

What methodological advances might improve PHYHD1 research in the future?

Emerging technologies and methodological innovations poised to advance PHYHD1 research include:

  • CRISPR-based tools for endogenous tagging:

    • Knock-in fluorescent tags to visualize endogenous PHYHD1 localization without antibody limitations

    • Implement CRISPR activation/inhibition systems for temporal control of PHYHD1 expression

    • Generate tissue-specific conditional knockouts to probe function in specific cell populations

  • Advanced proximity labeling methods:

    • Apply TurboID or miniTurbo systems for rapid biotin labeling of PHYHD1 interaction partners

    • Implement spatially restricted enzymatic tagging to map compartment-specific interactions

    • Use split-TurboID to detect specific protein-protein interactions in living cells

  • Structural biology approaches:

    • Apply cryo-EM to visualize PHYHD1 complexes with interacting partners

    • Use hydrogen-deuterium exchange mass spectrometry to map conformational changes during substrate binding

    • Implement AlphaFold2-based modeling to predict interaction interfaces with Aβ42

  • Improved antibody technologies:

    • Develop recombinant nanobodies against PHYHD1 for super-resolution microscopy

    • Generate isoform-specific antibodies to distinguish between the three alternatively spliced variants

    • Create phospho-specific antibodies to track activity-dependent modifications

  • Functional metabolomics:

    • Implement metabolic tracing to identify substrates of PHYHD1's dioxygenase activity

    • Use stable isotope labeling to track alpha-ketoglutarate consumption by PHYHD1

    • Apply untargeted metabolomics to identify novel metabolites altered by PHYHD1 activity

  • In vivo imaging advances:

    • Develop PET tracers targeting PHYHD1 for longitudinal studies in AD models

    • Implement two-photon microscopy with PHYHD1 activity sensors in mouse models

    • Apply spatial transcriptomics to map PHYHD1 expression patterns in intact brain tissue

These methodological advances will help overcome current limitations in studying this relatively understudied protein with potential significance in neurodegenerative disease mechanisms .

What are best practices for integrating PHYHD1 research into broader neurodegenerative disease studies?

When incorporating PHYHD1 research into comprehensive neurodegenerative disease studies, researchers should follow these evidence-based recommendations:

  • Standardize PHYHD1 detection across studies:

    • Validate at least two independent antibodies against different epitopes

    • Include both protein and mRNA quantification methods

    • Document antibody validation methods explicitly in publications

  • Contextualize PHYHD1 within molecular networks:

    • Analyze alongside other proteins in its interaction network (C4A/C4B, CD74, CTSS, CX3CR1)

    • Consider its relationship with APP processing and Aβ metabolism

    • Examine correlations with neuroinflammatory markers

  • Implement longitudinal experimental designs:

    • Track PHYHD1 expression changes across disease progression

    • Correlate with cognitive and pathological markers

    • Compare age-matched controls carefully to distinguish disease-specific from aging effects

  • Adopt multi-omic approaches:

    • Combine transcriptomic, proteomic, and metabolomic analyses

    • Implement spatial transcriptomics to map regional expression patterns

    • Correlate genomic variations in PHYHD1 with expression levels and disease risk

  • Establish causality through intervention studies:

    • Use conditional knockout/knockdown models with appropriate controls

    • Implement rescue experiments to confirm specificity

    • Develop small molecule modulators of PHYHD1 activity for pharmacological validation

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 2025 TheBiotek. All Rights Reserved.