APH1B Antibody

Shipped with Ice Packs
In Stock

Description

Key Applications

  • Western blot (WB): Detects APH1B in brain lysates and cell lines .

  • Immunoprecipitation (IP): Isolates γ-secretase complexes for functional assays .

  • Immunohistochemistry (IHC): Localizes APH1B in hippocampal and cortical neurons .

Disease Relevance

  • Alzheimer’s Disease: Elevated APH1B expression in blood correlates with amyloid-β deposition and cortical thinning in AD patients .

  • Synaptic Dysfunction: APH1B deficiency disrupts dendritic spine formation and long-term potentiation in mice, implicating its role in schizophrenia-related pathways .

Validated APH1B Antibodies

The following table summarizes widely used APH1B antibodies and their validation data:

SupplierCatalog No.TypeApplicationsReferences
Novus BiologicalsNBP1-98414PolyclonalWB, IHC
InvitrogenAPH1B-101APPolyclonalWB, IP, ELISA
Cusabio BiotechCSB-PA819900PolyclonalWB, ELISA
Antibodies.comA52753PolyclonalWB, IHC, ICC

Table: APH1B in Disease Models and Biomarkers

Study FocusKey FindingsSource
AD BiomarkersBlood APH1B levels correlate with amyloid-β and entorhinal cortex thinning .PMC8567578
Synaptic PhenotypesAPH1B knockout mice show reduced dendritic spines and impaired LTP .eLife02196
Aβ ModulationAph1BC−/− mice exhibit 50% lower Aβ in brain extracts .PMC2740474
γ-Secretase ActivationAPH1B mutations (e.g., L30F/T164A) enhance Aβ production in vitro .PMC8745412

Challenges and Considerations

  • Isoform Cross-Reactivity: APH1B antibodies must distinguish between APH1A/B/C isoforms due to overlapping structural domains .

  • Functional Heterogeneity: APH1B-containing γ-secretase complexes preferentially cleave substrates like Neuregulin-1 over APP or Notch in vivo .

  • Therapeutic Potential: Targeting APH1B-specific γ-secretase activity reduces Aβ plaques in AD mouse models without Notch-related toxicity .

Future Directions

Research priorities include developing isoform-specific inhibitors for APH1B to mitigate Aβ pathology in AD and clarifying its role in neurodevelopmental disorders. Structural studies of APH1B-γ-secretase complexes bound to substrates like Aβ46 are ongoing to refine drug design .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery timelines may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributor.
Synonyms
APH1B; PSFL; UNQ688/PRO1328; Gamma-secretase subunit APH-1B; APH-1b; Aph-1beta; Presenilin-stabilization factor-like
Target Names
Uniprot No.

Target Background

Function
APH1B is likely a subunit of the gamma-secretase complex. This complex functions as an endoprotease, cleaving integral membrane proteins such as Notch receptors and APP (amyloid-beta precursor protein) within the membrane. APH1B potentially serves as a stabilizing cofactor for the presenilin homodimer, facilitating the formation of a stable complex. Compared to APH1A, APH1B is likely present in a smaller proportion of gamma-secretase complexes.
Gene References Into Functions
  1. Research indicates that gamma-secretase complexes composed of presenilin 1 (PS1)/anterior-pharynx-defective protein 1 (Aph1b), presenilin 2 (PS2)/Aph1aL, PS2/Aph1aS, and PS2/anterior pharynx defective 1 homolog B (Aph1b) generate amyloid beta peptide (Abeta) with a higher Abeta42+Abeta43-to-Abeta40 (Abeta42(43)/Abeta40) ratio compared to other gamma-secretase complexes. PMID: 27608597
  2. Inhibition of gamma-secretase during repeated peripheral inflammation has been shown to prevent central amyloid-beta accumulation and cognitive dysfunction. PMID: 23665252
  3. Expression of APH-1b has been associated with increased amyloid beta peptide levels and gamma-secretase activity. PMID: 12763021
  4. Both APH-1a splice forms (APH-1aS and APH-1aL) and APH-1b are expressed in peripheral and neuronal cells. They form distinct, proteolytically active gamma-secretase complexes containing either one of the two presenilins. PMID: 15286082
  5. Dimeric (NCSTN/APH-1) and trimeric (NCSTN/APH-1/PS1) intermediates of the gamma-secretase complex assembly are retained within the endoplasmic reticulum (ER). Incorporation of the fourth binding partner (PEN-2) also occurs on immature NCSTN (APH-1). PMID: 15591316
  6. Knockdown of APH-1a, but not APH-1b, resulted in impaired maturation of nicastrin and reduced expression of presenilin 1, presenilin 2, and PEN-2 proteins. PMID: 15629423
  7. APH-1b variant protein is destabilized, and the fourth transmembrane domain plays a critical role in protein stability and function of APH-1. PMID: 15823552
  8. Collectively, these findings suggest that the three forms of APH-1 can be interchangeable within presenilin (PS) complexes and that the transmembrane GxxxG region is crucial for the stability of the APH-1 protein and the assembly of PS complexes. PMID: 16757808
  9. Data suggest that a cooperative mechanism involving APOE and APH-1b plays a role in the susceptibility to develop Alzheimer's disease (AD). PMID: 17466415
  10. A non-synonymous single nucleotide polymorphism (SNP) in the gamma-secretase component APH1B (Phe217Leu; rs1047552) has been significantly associated with premature coronary atherosclerosis in Dutch males. PMID: 18987747
  11. The SNP (Phe217Leu; rs1047552) exhibited a tendency for association with HIV-1 infection in a Xhosa indigenous South African Bantu study (P = 0.087) and showed significant association in a Caucasian Dutch study (P = 0.049). PMID: 19774691

Show More

Hide All

Database Links

HGNC: 24080

OMIM: 607630

KEGG: hsa:83464

STRING: 9606.ENSP00000261879

UniGene: Hs.511703

Protein Families
APH-1 family
Subcellular Location
Membrane; Multi-pass membrane protein.
Tissue Specificity
Weakly or not expressed in leukocytes, lung, placenta, small intestine, liver, kidney, spleen thymus, colon, skeletal muscle, heart and brain.

Q&A

What is APH1B and what role does it play in cellular function?

APH1B is one of several homologues of the Anterior Pharynx Defective 1 protein that, together with presenilin, PEN2, and nicastrin, forms the γ-secretase protein complex. This complex is responsible for cleaving transmembrane proteins including the amyloid precursor protein (APP) and Notch receptors. APH1B functions as a stabilizing cofactor for the presenilin homodimer, promoting formation of a stable γ-secretase complex . In mammals, three APH1 homologues have been identified: APH1A, APH1B, and in rodents, a third gene called APH1C that arose from duplication of the APH1B gene . While APH1A is more abundantly expressed than APH1B in human tissues and is considered the major isoform required for Notch signaling during embryogenesis, APH1B has been implicated in specific aspects of APP processing in selective brain regions .

What types of APH1B antibodies are available for research applications?

Multiple types of APH1B antibodies are available for various research applications. These include polyclonal antibodies derived from rabbit hosts that target specific amino acid sequences of the APH1B protein . Available antibodies may target different epitopes, such as amino acids 51-150 or 93-110 . Researchers can choose from various conjugated forms including those with fluorescent tags (AbBy Fluor® 594, AbBy Fluor® 488, AbBy Fluor® 647, FITC), enzyme conjugates (HRP), or affinity tags like biotin . These diverse formats enable applications ranging from Western blotting to immunofluorescence microscopy in both cultured cells and tissue sections .

What are the typical applications for APH1B antibodies in neuroscience research?

APH1B antibodies are utilized in multiple neuroscience research applications focused on understanding γ-secretase activity and Alzheimer's disease pathogenesis. Common applications include:

  • Western blotting (WB) to detect and quantify APH1B protein expression in brain tissue or cell lysates

  • Immunofluorescence (IF) in both cultured cells and paraffin-embedded tissue sections to visualize the localization and distribution of APH1B

  • Immunohistochemistry (IHC) in frozen or paraffin-embedded brain tissues to examine regional expression patterns

  • Analysis of γ-secretase activity through measurement of substrate processing (e.g., APP or Notch)

  • Investigation of associations between APH1B expression and neuroimaging biomarkers of Alzheimer's disease

How should researchers select the appropriate APH1B antibody for their specific application?

When selecting an APH1B antibody, researchers should consider multiple factors based on their experimental needs:

  • Epitope specificity: Determine whether the research question requires targeting specific regions of APH1B. For instance, antibodies targeting amino acids 51-150 versus 93-110 may have different recognition properties .

  • Species reactivity: Confirm the antibody's reactivity with the species being studied. Some APH1B antibodies show reactivity with mouse samples but are only predicted to react with human samples, while others have confirmed human reactivity .

  • Application compatibility: Verify that the antibody has been validated for the intended application. For example, some APH1B antibodies are validated for Western blotting, immunofluorescence, and immunohistochemistry, while others may have more limited validation .

  • Conjugation requirements: Select appropriate conjugates based on detection methods. Fluorophore-conjugated antibodies (AbBy Fluor® 594, 488, 647) are optimal for immunofluorescence, while HRP or biotin conjugates may be preferred for other applications .

  • Validation evidence: Review available validation data demonstrating specificity, including testing in knockout models where APH1B expression is absent .

What controls are essential when using APH1B antibodies to study γ-secretase function?

Implementing proper controls is critical for ensuring reliable results when studying γ-secretase function with APH1B antibodies:

  • Genetic controls: When available, include samples from APH1B knockout or knockdown models to confirm antibody specificity. Studies have utilized fibroblasts derived from APH1B knockout animals to validate antibody specificity and examine compensatory expression of other APH1 homologues .

  • Peptide competition: Use the immunizing peptide to block antibody binding in a parallel sample to demonstrate binding specificity.

  • Cross-reactivity assessment: Test the antibody against related proteins (APH1A, APH1C) to ensure it specifically recognizes APH1B. This is particularly important as these proteins share sequence similarity .

  • Functional assays: When studying γ-secretase activity, include positive controls such as known γ-secretase substrates (APP and Notch), and negative controls such as samples treated with γ-secretase inhibitors .

  • Tissue expression pattern controls: Compare antibody staining patterns to known expression patterns of APH1B in different tissues and brain regions. The brainstem and olfactory bulb show higher expression levels and may serve as positive control regions .

How can researchers quantitatively assess γ-secretase activity in relation to APH1B expression?

Several methodological approaches can be employed to quantitatively assess γ-secretase activity in relation to APH1B expression:

  • Western blot analysis of substrate accumulation: Measure the accumulation of APP C-terminal fragments (APP-CTFs), which accumulate when γ-secretase activity is reduced. Studies in APH1BC-deficient mice demonstrated a >2-fold accumulation of APP-CTF in brainstem and olfactory bulb regions .

  • Reporter gene assays: Utilize UAS-luciferase reporter systems with APP or Notch reporter constructs containing Gal4-VP16 sequences in their cytoplasmic domains. This approach enables direct comparison of processing efficiencies for different substrates .

  • Measurement of proteolytic products: Detect and quantify specific cleavage products such as NICD (Notch Intracellular Domain) or Aβ (amyloid-beta peptide) using antibodies specific to these products .

  • Expression correlation analyses: Examine correlations between APH1B expression levels and neuroimaging biomarkers such as entorhinal cortical thickness or global cortical amyloid-β deposition, as demonstrated in studies utilizing ADNI and AddNeuroMed cohorts .

How can APH1B antibodies be utilized to investigate differential γ-secretase complex composition in various tissues?

APH1B antibodies provide powerful tools for investigating tissue-specific γ-secretase complex composition:

  • Co-immunoprecipitation studies: APH1B antibodies can be used to isolate intact γ-secretase complexes from different tissues, followed by mass spectrometry or Western blotting to identify associated proteins. This approach can reveal tissue-specific complex compositions.

  • Comparative Western blotting: Quantitative Western blotting with antibodies against different γ-secretase components (APH1A, APH1B, presenilin, nicastrin, PEN2) across tissue samples can reveal differential expression ratios. Research has demonstrated that Aph1BC deficiency affected Psen1 and Pen2 steady-state levels most clearly in brainstem extracts .

  • Immunofluorescence co-localization: Multiple-label immunofluorescence studies combining APH1B antibodies with antibodies against other γ-secretase components can reveal spatial distribution patterns in tissues and subcellular compartments.

  • Cross-linking studies: Chemical cross-linking followed by immunoprecipitation with APH1B antibodies can stabilize transient interactions within the γ-secretase complex for analysis.

  • Sequential extraction protocols: Different solubilization conditions combined with APH1B immunodetection can reveal differential complex stability across tissues.

What experimental approaches can resolve contradictory findings regarding APH1B's role in Alzheimer's disease pathology?

Resolving contradictory findings regarding APH1B's role in Alzheimer's disease pathology requires sophisticated experimental approaches:

  • Cell-type specific analyses: Use APH1B antibodies in combination with cell-type markers to determine whether effects are specific to neurons, glia, or vascular cells. Single-cell transcriptomics data can be integrated with protein-level analyses.

  • Regional brain analysis: Systematically analyze APH1B expression and γ-secretase activity across brain regions relevant to AD pathology. Studies have found that APH1BC deficiency had differential effects on APP processing across brain regions, with the strongest effects observed in brainstem and olfactory bulb .

  • Longitudinal studies: Examine how APH1B expression changes during disease progression using animal models at different disease stages.

  • Patient stratification: Analyze APH1B expression in relation to genetic risk factors (e.g., APOE status) or biomarker profiles to identify potential subgroup-specific effects.

  • Integration of blood and brain analyses: Combined analysis of blood-based APH1B expression with neuroimaging biomarkers can reveal systemic relationships. Research has demonstrated that APH1B expression levels in blood are associated with entorhinal cortical thickness and global cortical amyloid-β deposition .

How can researchers distinguish between direct effects of APH1B modulation and compensatory mechanisms in experimental models?

Distinguishing direct effects from compensatory mechanisms requires careful experimental design:

  • Acute versus chronic modulation: Compare acute knockdown (e.g., siRNA) with chronic genetic deficiency to differentiate immediate effects from compensatory adaptations.

  • Temporal analysis: Examine changes in other γ-secretase components at multiple time points following APH1B modulation. Research has shown that knockout of APH1 genes did not lead to major compensatory up-regulation of other APH1 homologues in fibroblast models .

  • Dose-dependent effects: Use graded expression systems to determine whether effects scale with APH1B levels or exhibit threshold phenomena.

  • Rescue experiments: Test whether reintroduction of APH1B can reverse observed phenotypes, confirming direct rather than indirect effects.

  • Cross-species comparison: Compare findings across mouse, rat, and human systems to identify conserved versus species-specific effects. Studies have identified similar patterns of γ-secretase activity changes in rat models with diminished Aph1BC expression as observed in mice .

The following table summarizes observed effects of APH1B/C deficiency on APP processing in different brain regions:

Brain RegionAPP-CTF AccumulationStatistical SignificanceOther Effects
Brainstem>2-foldSignificantAltered Psen1 and Pen2 levels
Olfactory Bulb>2-foldSignificantNot specified
CerebellumMildSignificantNot specified
Other Brain RegionsMildNot significantNot specified

How can blood-based APH1B expression analysis be utilized for Alzheimer's disease biomarker development?

Recent research has revealed promising applications for blood-based APH1B expression analysis in Alzheimer's disease biomarker development:

What methodological considerations are critical when using APH1B antibodies for quantitative analysis in human samples?

Quantitative analysis of APH1B in human samples requires careful attention to methodological details:

  • Sample preparation standardization: Establish consistent protocols for blood collection, processing, and storage to minimize pre-analytical variability. Timing of sample collection and processing can significantly impact protein expression measurements.

  • Antibody validation in human samples: Verify antibody specificity in human tissues using peptide competition, multiple antibodies targeting different epitopes, and correlation with mRNA expression data.

  • Internal standards: Include recombinant APH1B protein standards at known concentrations to enable absolute quantification rather than relative comparisons.

  • Normalization strategy: Carefully select appropriate housekeeping proteins or global protein measurement approaches for normalization, as traditional housekeeping genes may exhibit variability in disease states.

  • Batch effect management: Implement randomization and batch correction strategies, particularly for large-scale studies involving multiple processing batches.

  • Clinical data integration: Collect comprehensive clinical data including medication history, as some medications might influence γ-secretase activity or APH1B expression.

How might differential targeting of APH1B-containing γ-secretase complexes inform therapeutic development for Alzheimer's disease?

The selective targeting of APH1B-containing γ-secretase complexes presents intriguing therapeutic possibilities:

  • Reduced toxicity potential: Unlike APH1A-deficient mice that exhibit lethal developmental phenotypes, APH1BC-deficient mice survive into adulthood, suggesting that targeting APH1B-containing complexes might achieve partial γ-secretase inhibition with fewer adverse effects . This implies the theoretical possibility that targeting specific γ-secretase subunit combinations could yield less toxic drugs than currently available general inhibitors of γ-secretase activity.

  • Region-specific modulation: The differential effects of APH1BC deficiency across brain regions suggests that targeting these complexes might modulate APP processing in a region-specific manner, potentially providing more precise therapeutic effects .

  • Substrate selectivity: Different γ-secretase complexes may exhibit differential substrate preferences. Therapeutic approaches targeting APH1B-containing complexes might achieve selective reduction in amyloid production while preserving essential Notch signaling functions mediated primarily by APH1A-containing complexes .

  • Combined biomarker-therapeutic approach: Blood APH1B expression levels could potentially identify patients most likely to benefit from therapies targeting APH1B-containing γ-secretase complexes, enabling a precision medicine approach .

  • Behavioral considerations: Studies in rat models with diminished APH1BC expression have revealed neurobehavioral abnormalities, including increased sensitivity to apomorphine and changes in prepulse and latent inhibition . These observations indicate that comprehensive psychopharmacological evaluation would be essential for any therapeutic approach targeting APH1B.

What optimization strategies should researchers employ when using APH1B antibodies for immunofluorescence applications?

Optimizing immunofluorescence protocols with APH1B antibodies requires attention to several technical details:

  • Fixation method selection: Compare aldehyde-based (paraformaldehyde, glutaraldehyde) versus alcohol-based (methanol, ethanol) fixation methods, as membrane proteins like APH1B may exhibit differential epitope accessibility depending on fixation approach.

  • Antigen retrieval optimization: Test multiple antigen retrieval methods (heat-induced epitope retrieval in citrate or Tris buffers at various pH values, or enzymatic retrieval) to maximize signal while preserving tissue morphology.

  • Blocking protocol refinement: Optimize blocking conditions using combinations of serum, BSA, and detergents to minimize background while preserving specific signals. The search results indicate that some APH1B antibodies are provided in buffers containing BSA, suggesting this may help stabilize antibody-epitope interactions .

  • Signal amplification consideration: For low abundance targets, evaluate signal amplification methods such as tyramide signal amplification or secondary antibody polymers.

  • Multi-label protocol development: When combining APH1B antibodies with other markers, carefully plan the staining sequence, antibody combinations, and fluorophore selection to avoid cross-reactivity and spectral overlap.

  • Confocal microscopy parameters: Optimize confocal microscopy settings including pinhole size, detector gain, and laser power to achieve optimal signal-to-noise ratio while avoiding photobleaching.

What are the critical parameters for successful Western blotting detection of APH1B in complex tissue samples?

Successful Western blotting detection of APH1B requires optimization of several parameters:

  • Sample preparation: Optimize tissue lysis conditions using buffers containing appropriate detergents (e.g., CHAPS, Triton X-100) for efficient solubilization of membrane-associated γ-secretase complexes. Consider using protease inhibitor cocktails to prevent degradation.

  • Protein loading optimization: Determine optimal protein loading amounts through titration experiments. APH1B may require higher loading amounts in tissues with lower expression levels.

  • Gel percentage selection: Select appropriate acrylamide percentage based on APH1B's molecular weight (~25-30 kDa) to achieve optimal resolution. Higher percentage gels (12-15%) typically provide better resolution for lower molecular weight proteins.

  • Transfer conditions: Optimize transfer conditions (voltage, time, buffer composition) for efficient transfer of hydrophobic membrane proteins. Semi-dry versus wet transfer methods may yield different results.

  • Antibody concentration titration: Perform antibody dilution series to determine optimal concentration that maximizes specific signal while minimizing background. Research articles have successfully used APH1B antibodies for Western blotting of fibroblast and brain tissue samples .

  • Incubation conditions: Optimize primary antibody incubation temperature and duration (4°C overnight versus room temperature for shorter periods) to enhance specific binding.

  • Detection system selection: Compare chemiluminescence, near-infrared fluorescence, and chromogenic detection systems to determine the most appropriate method for your specific application and sensitivity requirements.

What emerging applications of APH1B antibodies might advance our understanding of neurodegenerative diseases?

Several emerging applications of APH1B antibodies hold promise for advancing neurodegenerative disease research:

  • Single-cell proteomics: Combining APH1B antibodies with single-cell analysis techniques could reveal cell-type specific variations in γ-secretase complex composition and function within heterogeneous brain tissues.

  • In vivo imaging: Development of near-infrared fluorophore-conjugated APH1B antibodies or antibody fragments for non-invasive imaging might enable longitudinal studies of γ-secretase activity in animal models.

  • Proximity labeling approaches: Using APH1B antibodies in conjunction with proximity labeling techniques (BioID, APEX) could identify novel interaction partners specific to different cellular contexts or disease states.

  • Spatial transcriptomics integration: Combining APH1B immunohistochemistry with spatial transcriptomics could reveal relationships between APH1B protein expression, γ-secretase activity, and local transcriptional networks.

  • Extracellular vesicle analysis: Investigating APH1B in extracellular vesicles might provide insights into cell-to-cell transmission of γ-secretase components and their potential role in disease propagation.

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.