APPL2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to APPL2 Antibody

The APPL2 antibody is a specialized research tool designed to detect and study the adaptor protein APPL2 (Adaptor Protein, Phosphotyrosine interaction, PH domain, and Leucine zipper containing 2), a multifunctional regulator of cellular signaling pathways. This antibody enables researchers to investigate APPL2's roles in processes such as glucose metabolism, immune response, intracellular trafficking, and cytoskeletal dynamics .

Role and Function of APPL2

APPL2 is a BAR domain-containing protein that interacts with membrane receptors, GTPases, and signaling complexes. Key functions include:

  • Regulation of Insulin Secretion: APPL2 modulates glucose-stimulated insulin secretion (GSIS) in pancreatic β-cells by promoting F-actin depolymerization via Rac1 activation .

  • Immune Modulation: APPL2 suppresses TLR4-mediated inflammatory responses by controlling NF-κB nuclear translocation and cytokine secretion .

  • Metabolic Signaling: Antagonizes APPL1 to regulate adiponectin and insulin signaling pathways, influencing glucose uptake and thermogenesis .

Key Applications and Performance

ApplicationTested SpeciesRecommended DilutionKey Studies
Western Blot (WB)Human, Mouse, Rat1:500–1:2000Detection of APPL2 in pancreatic islets, neurons, and cancer cell lines .
Immunohistochemistry (IHC)Human, Mouse1:100–1:1000Localization in brain, pancreas, and melanoma tissues .
Immunofluorescence (IF)Human1:50–1:500Visualization of APPL2 in endosomal membranes and nuclei .
ELISAHumanNot specifiedQuantification in serum and cell lysates .

Glucose-Stimulated Insulin Secretion (GSIS)

  • APPL2 deficiency in β-cells impairs both first- and second-phase insulin secretion by disrupting F-actin remodeling via RacGAP1 inhibition .

  • Validated using phalloidin staining and live-cell imaging in pancreatic islets from APPL2 knockout mice .

TLR4 Signaling in Inflammation

  • APPL2 suppresses LPS-induced NF-κB activation and pro-inflammatory cytokine release in macrophages, as shown via co-immunoprecipitation and knockdown experiments .

Cancer and Neurodegenerative Disease Links

  • Overexpression detected in pancreatic cancer (BxPC-3 cells) and glioblastoma using IHC .

  • Implicated in amyloid precursor protein (APP) processing, with relevance to Alzheimer’s disease .

Comparative Analysis

SupplierCatalog No.HostReactivityImmunogenValidation
Abcamab154661RabbitHumanRecombinant fragment (aa 450–C-terminus)WB, IHC-P, ICC/IF
Proteintech14294-1-APRabbitHuman, MouseFull-length fusion proteinWB, IHC, IF/ICC
Assay GenieCAB14590RabbitHuman, Mouse, RatRecombinant fragment (aa 1–300)WB, ELISA
Novus BiologicalsNBP1-89029RabbitHuman, MouseSynthetic peptide (Leu37–Ser680)IHC, IF, Flow Cytometry

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery information.
Synonyms
Adapter protein containing PH domain antibody; Adapter protein containing PH domain PTB domain and leucine zipper motif 2 antibody; adaptor protein; phosphotyrosine interaction; PH domain and leucine zipper containing 2 antibody; APLP2 antibody; Appl2 antibody; DCC interacting protein 13 beta antibody; DCC-interacting protein 13-beta antibody; Dip13 beta antibody; Dip13-beta antibody; DP13B_HUMAN antibody; PTB domain and leucine zipper motif 2 antibody
Target Names
APPL2
Uniprot No.

Target Background

Function
APPL2 is a multifunctional adapter protein that interacts with various membrane receptors, nuclear factors, and signaling proteins to regulate diverse cellular processes, including cell proliferation, immune response, endosomal trafficking, and cell metabolism. It plays a crucial role in regulating signaling pathways involved in cell proliferation through its interaction with RAB5A and subunits of the NuRD/MeCP1 complex. APPL2 participates in immune response by modulating phagocytosis, inflammatory, and innate immune responses. In macrophages, it enhances Fc-gamma receptor-mediated phagocytosis through interaction with RAB31, leading to activation of the PI3K/Akt signaling pathway. In response to LPS, APPL2 modulates inflammatory responses by playing a key role in the regulation of TLR4 signaling and in the nuclear translocation of RELA/NF-kappa-B p65, ultimately impacting the secretion of pro- and anti-inflammatory cytokines. APPL2 also functions as a negative regulator of the innate immune response by inhibiting the AKT1 signaling pathway through the formation of a complex with APPL1 and PIK3R1. APPL2 is involved in the endosomal trafficking of TGFBR1 from the endosomes to the nucleus. Furthermore, APPL2 plays a role in cell metabolism by regulating adiponectin and insulin signaling pathways as well as adaptive thermogenesis. In muscle, APPL2 negatively regulates adiponectin-stimulated glucose uptake and fatty acid oxidation by inhibiting adiponectin signaling through APPL1 sequestration, antagonizing APPL1 action. In muscles, it negatively regulates insulin-induced plasma membrane recruitment of GLUT4 and glucose uptake through interaction with TBC1D1. APPL2 contributes to cold and diet-induced adaptive thermogenesis by activating ventromedial hypothalamus (VMH) neurons through AMPK inhibition, which enhances sympathetic outflow to subcutaneous white adipose tissue (sWAT), sWAT beiging, and cold tolerance. APPL2 is also involved in other signaling pathways, including Wnt/beta-catenin, HGF, and glucocorticoid receptor signaling. It functions as a positive regulator of beta-catenin/TCF-dependent transcription through direct interaction with RUVBL2/reptin, resulting in the relief of RUVBL2-mediated repression of beta-catenin/TCF target genes by modulating the interactions within the beta-catenin-reptin-HDAC complex. APPL2 may affect adult neurogenesis in the hippocampus and olfactory system via regulating the sensitivity of the glucocorticoid receptor. Finally, APPL2 is required for fibroblast migration through HGF cell signaling.
Gene References Into Functions
  1. Studies have shown that the suppressive effect of OCC-1 RNA on the transcription level of the APPL2 gene provides a putative colorectal neoplasm progression index. PMID: 27986894
  2. Research has demonstrated that signal transducing adaptor proteins APPL1 and APPL2 are essential for TGFbeta-induced nuclear translocation of TGFbeta type I receptor (TbetaRI)-ICD and for cancer cell invasiveness in prostate and breast cancer cell lines. PMID: 26583432
  3. ATM serves as the central modulator of APPL-mediated effects on radiosensitivity and DNA repair. PMID: 24763056
  4. Data indicate that APPL2(PH) binding to the BAR domain and Reptin is mutually exclusive, which regulates the nucleocytoplasmic shuttling of Reptin. PMID: 23891720
  5. Findings suggest that the C-APPL1/A-APPL2 allele combination is associated with non-alcoholic fatty liver disease occurrence, with a more severe hepatic steatosis grade and a reduced adiponectin cytoprotective effect on the liver. PMID: 23977033
  6. Data indicate that a high level of APPL2 protein might enhance glioblastoma growth by maintaining low expression levels of genes responsible for cell death induction. PMID: 22989406
  7. Analysis of APPL1 and APPL2 proteins and their interaction with Rab has been conducted. PMID: 23055524
  8. Significant evidence of association with overweight/obesity has been found for rs2272495 and rs1107756. The rs2272495 C allele and rs1107756 T allele both conferred a higher risk of being overweight and obese. PMID: 22462604
  9. Genetic variations in APPL1/2 may be associated with CAD risk in T2DM in the Chinese population. PMID: 22340213
  10. Data suggest that while annexin A2 is not an exclusive marker of APPL1/2 endosomes, it plays a significant role in membrane recruitment of APPL proteins, functioning in parallel to Rab5. PMID: 21645192
  11. Significant fluorescence resonance energy transfer between APPL minimal BAR domain FRET pairs has been observed. PMID: 20814572
  12. Research has identified a pathway directly linking the small GTPase Rab5, a key regulator of endocytosis, to signal transduction and mitogenesis via APPL1 and APPL2, two Rab5 effectors. PMID: 15016378
  13. Findings suggest a role for APPL1 and APPL2 proteins as dynamic scaffolds that modulate RAB5-associated signaling endosomal membranes through their ability to undergo domain-mediated oligomerization, membrane targeting, and phosphoinositide binding. PMID: 18034774
  14. APPL proteins exert their stimulatory effects on beta-catenin/TCF-dependent transcription by decreasing the activity of a Reptin-containing repressive complex. PMID: 19433865

Show More

Hide All

Database Links

HGNC: 18242

OMIM: 606231

KEGG: hsa:55198

STRING: 9606.ENSP00000258530

UniGene: Hs.506603

Involvement In Disease
A chromosomal aberration involving APPL2/DIP13B is found in patients with chromosome 22q13.3 deletion syndrome. Translocation t(12;22)(q24.1;q13.3) with SHANK3/PSAP2 (PubMed:11431708).
Subcellular Location
Early endosome membrane; Peripheral membrane protein. Nucleus. Cell membrane. Endosome membrane. Cytoplasm. Cytoplasmic vesicle, phagosome. Cell projection, ruffle. Cell projection, ruffle membrane. Cell membrane. Cytoplasmic vesicle, phagosome membrane.
Tissue Specificity
High levels in brain, heart, kidney and skeletal muscle.

Q&A

What is APPL2 and why is it important in scientific research?

APPL2 (Adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 2) is a protein that plays significant roles in cell cycle regulation and carbohydrate metabolism and homeostasis. In humans, the canonical APPL2 protein consists of 664 amino acid residues with a molecular mass of approximately 74.5 kDa. Up to three different isoforms have been reported for this protein . APPL2 is notably expressed in the brain, heart, kidney, and skeletal muscle tissues, making it an important target for studies involving these organ systems. The protein is also known by several synonyms, including DCC-interacting protein 13-beta (DIP13 beta), adapter protein containing PH domain, PTB domain and leucine zipper motif 2, and DIP13B . Given its diverse functions and tissue distribution, APPL2 antibodies are valuable tools for investigating various physiological and pathological processes.

What types of APPL2 antibodies are available for research?

Both polyclonal and monoclonal antibodies against APPL2 are available for research purposes. Polyclonal antibodies, such as rabbit polyclonal anti-APPL2 antibodies, are commonly used in various applications . These antibodies recognize multiple epitopes on the APPL2 protein, potentially providing higher sensitivity but possibly lower specificity compared to monoclonal antibodies. The choice between polyclonal and monoclonal antibodies depends on the specific research application and the desired balance between sensitivity and specificity. For instance, polyclonal antibodies are often preferred for applications requiring strong signal detection, while monoclonal antibodies may be better suited for applications requiring high specificity .

What are the common applications for APPL2 antibodies in research?

APPL2 antibodies are commonly used in several research applications, including:

  • Western Blot (WB): For detecting and quantifying APPL2 protein in tissue or cell lysates

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of APPL2 in solution

  • Immunohistochemistry (IHC): For visualizing APPL2 distribution in tissue sections

  • Immunocytochemistry/Immunofluorescence (ICC-IF): For examining APPL2 localization within cells

Western Blot is particularly widely used for APPL2 detection, as it allows researchers to determine the molecular weight of the detected protein and confirm its identity. ELISA offers quantitative measurements of APPL2 levels in biological samples. Immunohistochemical and immunofluorescence techniques provide valuable information about the spatial distribution of APPL2 within tissues and cells, respectively.

How should I optimize antigen retrieval for APPL2 immunohistochemistry?

Optimizing antigen retrieval is crucial for successful APPL2 immunohistochemistry, especially when working with formalin-fixed, paraffin-embedded tissues. The choice of antigen retrieval method depends on the specific anti-APPL2 antibody being used, with monoclonal antibodies typically requiring more rigorous retrieval than polyclonal antibodies .

Two primary methods can be considered:

  • Heat-Induced Epitope Retrieval (HIER): This is often the preferred method for many antibodies. For APPL2 detection, citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) can be used, with the specific buffer optimized for your particular antibody. The tissue sections should be heated to 95-100°C for 15-20 minutes and then allowed to cool slowly to room temperature.

  • Proteolytic-Induced Epitope Retrieval (PIER): Enzymes such as trypsin, proteinase K, or pronase can be used, but this method may be less controlled and could potentially damage some epitopes. PIER works through protein digestion, which is non-specific, so careful optimization is required .

It's important to note that approximately 85% of antigens fixed in formalin require some type of antigen retrieval to optimize immunoreactivity . When developing a new protocol, testing both methods on control tissues is advisable to determine which provides the best signal-to-noise ratio for your specific anti-APPL2 antibody.

What controls should be included when using APPL2 antibodies?

Proper controls are essential for validating results obtained with APPL2 antibodies:

  • Positive Controls: Tissues known to express APPL2, such as brain, heart, kidney, or skeletal muscle samples, should be included to confirm antibody activity . These tissues should be processed identically to the experimental samples.

  • Negative Controls: These should include:

    • Omission of primary antibody (substituting with antibody diluent)

    • Isotype controls (using non-specific antibodies of the same isotype)

    • Tissue known not to express APPL2 or tissue from APPL2 knockout models if available

  • Absorption Controls: Pre-incubating the antibody with purified APPL2 protein should eliminate specific staining.

  • Species Compatibility Controls: When working with non-human samples, confirm cross-reactivity with the species of interest, as APPL2 orthologs have been reported in mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken species .

Additionally, it's important to validate each new lot of antibody, as there can be lot-to-lot variations that affect performance. Documentation of all control results is essential for publication and reproducibility purposes.

How can I quantify APPL2 expression in tissue samples?

Quantification of APPL2 expression can be approached through several methodologies:

  • Western Blot Densitometry:

    • Run protein samples alongside molecular weight markers

    • Detect APPL2 using validated antibodies

    • Use image analysis software to measure band intensities

    • Normalize to housekeeping proteins (e.g., GAPDH, β-actin)

  • Immunohistochemistry Quantification:

    • Use standardized staining protocols

    • Capture digital images under consistent conditions

    • Apply quantitative image analysis to measure:

      • Percentage of positive cells

      • Staining intensity (often on a 0-3 scale)

      • H-score (combining percentage and intensity)

  • ELISA-Based Quantification:

    • Develop standard curves using purified APPL2 protein

    • Measure absorbance values of unknown samples

    • Calculate concentration based on standard curve

For immunohistochemical quantification, consider the following table for standardized scoring:

ScoreStaining IntensityPercentage of Positive Cells
0Negative0%
1Weak1-25%
2Moderate26-50%
3Strong51-75%
4Very Strong76-100%

The H-score can be calculated as: ∑(intensity score × percentage of positive cells), resulting in a range from 0 to 400.

When reporting results, clearly document the quantification method, antibody details, and scoring system to ensure reproducibility .

How can I distinguish between different APPL2 isoforms in my experiments?

Distinguishing between the three reported APPL2 isoforms requires careful experimental design and selection of appropriate antibodies and techniques:

  • Antibody Selection: Choose antibodies raised against epitopes that are unique to specific isoforms or present in all isoforms, depending on your research needs. Contact antibody manufacturers for detailed epitope information to determine which isoforms each antibody can detect.

  • Western Blot Analysis: This technique can separate isoforms based on molecular weight differences. Use high-resolution SDS-PAGE (10-12% gels) for optimal separation of isoforms. The canonical human APPL2 protein has a reported mass of 74.5 kDa, while other isoforms may have different molecular weights .

  • PCR-Based Methods: Design primers specific to each isoform to detect isoform-specific mRNA expression:

    • Standard RT-PCR for qualitative analysis

    • qRT-PCR for quantitative comparison

    • Digital PCR for absolute quantification

  • Mass Spectrometry: For definitive identification, immunoprecipitate APPL2 from your samples and analyze the purified protein by mass spectrometry to identify isoform-specific peptides.

  • Recombinant Protein Controls: Express each APPL2 isoform recombinantly to serve as positive controls for antibody validation and to confirm the molecular weight of each isoform.

What are the best approaches for studying APPL2 interactions with other proteins?

To study APPL2 interactions with other proteins, consider these methodological approaches:

  • Co-Immunoprecipitation (Co-IP):

    • Use anti-APPL2 antibodies to pull down APPL2 and its binding partners

    • Verify interactions by western blotting with antibodies against suspected binding partners

    • Consider crosslinking to stabilize transient interactions

    • Include appropriate negative controls (IgG or unrelated antibody)

  • Proximity Ligation Assay (PLA):

    • Allows visualization of protein-protein interactions in situ

    • Requires antibodies against both APPL2 and potential binding partners raised in different species

    • Provides spatial resolution of interactions within cells or tissues

  • FRET/BRET Analysis:

    • Tag APPL2 and potential binding partners with appropriate fluorophores or luciferase

    • Measure energy transfer as an indicator of protein proximity

    • Allows real-time monitoring of dynamic interactions

  • Yeast Two-Hybrid Screening:

    • Use APPL2 as bait to identify novel interaction partners

    • Follow up with validation using the methods above

    • Consider domain-specific constructs to map interaction surfaces

  • Mass Spectrometry-Based Approaches:

    • Immunoprecipitate APPL2 complexes and identify binding partners by LC-MS/MS

    • SILAC or TMT labeling can provide quantitative information about interaction dynamics

    • Crosslinking MS can provide structural insights into the interaction interface

Each method has advantages and limitations, so using multiple complementary approaches provides the most reliable results. Consider the biological context of the interaction (e.g., cell type, stimulus conditions) when designing experiments.

How can advanced genetic techniques be used to study APPL2 function?

Advanced genetic techniques provide powerful tools for studying APPL2 function:

  • CRISPR/Cas9 Gene Editing:

    • Generate APPL2 knockout cell lines or animal models

    • Create point mutations to study specific domains or post-translational modification sites

    • Develop knockin models with fluorescent tags for live imaging

    • Design conditional knockout models for tissue-specific studies

  • RNAi and Antisense Technologies:

    • siRNA for transient APPL2 knockdown in cell culture

    • shRNA for stable knockdown via lentiviral delivery

    • Antisense oligonucleotides for in vivo applications

    • APPL2-targeting miRNAs to study endogenous regulation

  • Overexpression Systems:

    • Transfect cells with APPL2 expression constructs

    • Use inducible promoters for temporal control

    • Create domain deletion mutants to map functional regions

    • Express APPL2 fused to affinity tags for purification or detection

  • Single-Cell Analysis:

    • Examine APPL2 expression in heterogeneous tissues

    • Correlate APPL2 levels with cell states or phenotypes

    • Identify cell populations where APPL2 is most active

  • Transgenic Animal Models:

    • Study tissue-specific functions using Cre-loxP systems

    • Analyze phenotypic consequences of APPL2 modulation

    • Evaluate APPL2 function in disease models

When designing genetic manipulation experiments, consider:

  • The presence of APPL2 orthologs in your model organism of interest (APPL2 orthologs have been reported in mouse, rat, bovine, frog, zebrafish, chimpanzee and chicken species)

  • Potential compensation by related proteins

  • Off-target effects of genetic manipulation tools

  • Appropriate controls (scrambled siRNA, empty vectors, etc.)

How can I troubleshoot false positive or negative results with APPL2 antibodies?

Troubleshooting false results with APPL2 antibodies requires systematic evaluation of experimental conditions:

For False Positives:

  • Non-specific Binding:

    • Implement more stringent blocking (5% BSA or normal serum)

    • Increase washing duration and frequency

    • Optimize antibody concentration (perform titration experiments)

    • Consider using monoclonal antibodies for greater specificity

  • Cross-Reactivity:

    • Validate antibody specificity using APPL2 knockout/knockdown controls

    • Perform peptide competition assays

    • Test antibody against recombinant APPL2 and related proteins

    • Verify results with a second antibody targeting a different epitope

  • Background Staining:

    • Block endogenous peroxidase (for IHC) or biotin (for avidin-biotin systems)

    • Use polymer-based detection systems instead of avidin-biotin

    • Consider autofluorescence quenching for immunofluorescence

    • Optimize counterstaining procedures

For False Negatives:

  • Epitope Masking:

    • Test different antigen retrieval methods (heat vs. enzymatic)

    • Adjust fixation protocols (reduced fixation time)

    • Try different antibodies targeting different epitopes

    • Consider native conditions for immunoprecipitation

  • Antibody Issues:

    • Verify antibody activity with positive controls

    • Check for antibody degradation (proper storage conditions)

    • Validate antibody lot against previously working lots

    • Consider species cross-reactivity if working with non-human samples

  • Detection System:

    • Use more sensitive detection methods (signal amplification)

    • Extend incubation times for primary antibody

    • Test different detection antibodies/systems

    • Optimize chromogen development time

Systematic documentation of troubleshooting experiments will help identify the source of problems and establish reliable protocols for future experiments.

How should I interpret contradictory APPL2 expression data from different antibodies or techniques?

When faced with contradictory APPL2 expression data, a methodical approach to resolution includes:

  • Critical Evaluation of Antibodies:

    • Compare epitopes targeted by different antibodies

    • Assess antibody validation data from manufacturers

    • Check literature for reports of similar discrepancies

    • Consider whether antibodies detect different isoforms

  • Technical Considerations:

    • Different techniques have varying sensitivity thresholds

    • Western blot detects denatured protein; IHC/IF may detect native conformation

    • RNA-based methods (qPCR) measure transcription, not protein levels

    • Post-translational modifications may affect antibody recognition

  • Biological Variables:

    • Cell/tissue type differences in APPL2 expression

    • Developmental stage or disease state effects

    • Subcellular localization changes under different conditions

    • Consider potential splice variants or isoforms

  • Resolution Strategies:

    • Validate findings with orthogonal techniques

    • Use genetic approaches (siRNA, CRISPR) to confirm specificity

    • Employ mass spectrometry for definitive protein identification

    • Consider multiple antibodies targeting different epitopes

  • Data Integration:

    • Weigh evidence based on technical rigor of each method

    • Consider biological plausibility of each result

    • Evaluate whether discrepancies reveal novel biology

    • Be transparent about contradictions in publications

Remember that contradictory data often leads to new insights. Instead of dismissing discrepancies, investigate them thoroughly as they may reveal important biological complexity about APPL2 regulation or function.

What are the latest methodological advances in using antibodies for studying APPL2 in complex biological systems?

Recent methodological advances have expanded the capabilities for studying APPL2 in complex biological systems:

  • Multiplex Immunofluorescence and Imaging:

    • Simultaneous detection of APPL2 with multiple proteins

    • Spectral unmixing for distinguishing overlapping fluorophores

    • Cyclic immunofluorescence for detecting >40 proteins on a single specimen

    • Advanced microscopy (STORM, PALM, STED) for super-resolution imaging of APPL2 localization

  • Tissue Microarray Technology:

    • Allows screening of hundreds of tissues simultaneously for APPL2 expression

    • Enables high-throughput comparative studies across tissue types or disease states

    • Reduces variability by processing all samples under identical conditions

  • In Situ Proximity Ligation:

    • Detects protein-protein interactions involving APPL2 in fixed tissues

    • Visualizes modifications (phosphorylation, ubiquitination) on APPL2

    • Provides spatial context for molecular interactions

  • Mass Cytometry (CyTOF):

    • Metal-tagged antibodies for high-dimensional analysis

    • Simultaneous measurement of APPL2 with dozens of other proteins

    • Single-cell resolution without spectral overlap limitations

  • Mimetic Antibody Design:

    • Computational design of antibodies with enhanced specificity

    • Genetic algorithms to optimize molecular recognition

    • Development of new structural motifs for improved binding

  • Advanced Bioinformatics Integration:

    • Correlation of antibody-based data with genomic/proteomic datasets

    • Machine learning approaches for pattern recognition in complex datasets

    • Systems biology modeling of APPL2 in cellular networks

These advances enable more comprehensive understanding of APPL2 biology by providing context beyond simple expression patterns, revealing functional interactions, modification states, and dynamic behaviors in complex tissues and organisms.

How does APPL2 antibody staining differ between normal and pathological tissue samples?

Understanding the differential staining patterns of APPL2 in normal versus pathological tissues requires careful analysis and standardized protocols:

  • Normal Tissue Distribution:

    • APPL2 shows notable expression in brain, heart, kidney, and skeletal muscle tissues

    • Expression patterns may vary by cell type within these tissues

    • Subcellular localization may be predominantly cytoplasmic with possible membrane association

    • Baseline expression levels should be established for each tissue of interest

  • Pathological Alterations:

    • Changes may occur in:

      • Expression level (increased or decreased intensity)

      • Subcellular localization (nuclear translocation, membrane redistribution)

      • Pattern of expression (focal vs. diffuse)

      • Cell type specificity (altered expression in specific cell populations)

  • Quantification Approaches:

    • Digital image analysis with standardized acquisition parameters

    • Scoring systems that account for both intensity and distribution

    • Comparison of staining patterns using tissue microarrays

    • Correlation with other molecular markers

  • Interpretation Considerations:

    • Distinguish between adaptive responses and pathological changes

    • Consider context of APPL2's role in cell cycle and metabolism

    • Evaluate correlation with clinical parameters and outcomes

    • Assess whether changes are specific to particular disease subtypes

When comparing normal and pathological samples, standardization of all preanalytical variables is crucial, including sample procurement, fixation duration, tissue processing, and antigen retrieval methods . These factors can significantly impact staining results and lead to erroneous interpretations if not properly controlled.

What are the best practices for using APPL2 antibodies in comparative studies across species?

When conducting comparative studies of APPL2 across species, consider the following best practices:

  • Antibody Selection and Validation:

    • Choose antibodies raised against conserved epitopes

    • Verify cross-reactivity with each species of interest

    • Consider that APPL2 orthologs have been reported in mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken species

    • Validate each antibody against positive and negative controls from each species

  • Sequence Alignment Analysis:

    • Perform bioinformatic analysis of APPL2 sequences across target species

    • Identify conserved and divergent regions

    • Predict epitopes that may be affected by species-specific variations

    • Design experiments based on known sequence homology

  • Technical Standardization:

    • Use identical protocols for all species when possible

    • Adjust fixation parameters based on tissue characteristics

    • Optimize antigen retrieval specifically for each species/tissue combination

    • Process samples in parallel to minimize batch effects

  • Controls and Normalization:

    • Include species-specific positive and negative controls

    • Use housekeeping proteins conserved across species for normalization

    • Consider tissue-matched controls from each species

    • Include recombinant APPL2 proteins from each species when available

  • Data Interpretation:

    • Consider evolutionary context when interpreting differences

    • Distinguish between true biological differences and technical variations

    • Account for differences in tissue architecture across species

    • Be cautious about functional interpretations based solely on expression patterns

This methodical approach ensures that observed differences reflect true biological variation rather than technical artifacts, enabling meaningful cross-species comparisons of APPL2 biology.

How might emerging antibody technologies improve APPL2 detection and functional studies?

Emerging antibody technologies offer significant potential for advancing APPL2 research:

  • Designer Recombinant Antibodies:

    • Synthetically designed antibodies with enhanced specificity

    • Engineered to recognize specific APPL2 isoforms or post-translational modifications

    • Consistent production without batch-to-batch variation

    • Development using genetic algorithms and computational design approaches

  • Nanobodies and Single-Domain Antibodies:

    • Smaller size allows access to epitopes inaccessible to conventional antibodies

    • Improved penetration in tissue sections and live cells

    • Potential for intracellular expression to track APPL2 in living systems

    • Reduced immunogenicity for in vivo applications

  • Optogenetic Antibody Systems:

    • Light-activatable antibodies for spatiotemporal control

    • Allows precise manipulation of APPL2 function in specific subcellular regions

    • Enables real-time studies of APPL2's dynamic interactions and functions

    • Combines detection with functional perturbation

  • Antibody-Drug Conjugates for Functional Studies:

    • Targeted delivery of functional modulators to APPL2-expressing cells

    • Enables selective inhibition or activation of APPL2-dependent pathways

    • Allows precise manipulation of APPL2 function in specific cell populations

  • Multiplexed Detection Systems:

    • Simultaneous visualization of APPL2 with dozens of other proteins

    • Integration with single-cell sequencing for correlative multi-omics

    • Advanced computational analysis to identify APPL2-associated molecular networks

    • Spatial transcriptomics combined with APPL2 protein detection

These emerging technologies promise to transform APPL2 research from descriptive studies of expression patterns to functional investigations of its roles in complex biological systems, potentially revealing new therapeutic targets related to APPL2's functions in metabolism and cell cycle regulation.

What are the most promising computational approaches for optimizing APPL2 antibody design and validation?

Computational approaches are revolutionizing antibody design and validation, with several promising applications for APPL2 research:

  • Epitope Prediction and Optimization:

    • Machine learning algorithms to identify highly specific and accessible epitopes

    • Structural modeling to predict epitope exposure in native APPL2 protein

    • Comparative sequence analysis to identify conserved epitopes for cross-species studies

    • Prediction of post-translational modification sites that might affect antibody binding

  • Genetic Algorithm-Based Design:

    • Rapid in silico optimization of antibody-antigen interactions

    • Design of mimetic antibodies with enhanced specificity for APPL2

    • Computational screening of large antibody libraries against APPL2 structural models

    • Identification of new structural motifs optimized for APPL2 recognition

  • Molecular Dynamics Simulations:

    • Modeling of antibody-APPL2 binding kinetics and thermodynamics

    • Prediction of conformational changes that might affect epitope accessibility

    • Optimization of antibody complementarity-determining regions (CDRs)

    • Virtual screening of antibody candidates before experimental production

  • Cross-Reactivity Assessment:

    • In silico prediction of potential cross-reactivity with related proteins

    • Identification of unique APPL2 sequences for highly specific antibody generation

    • Prediction of species cross-reactivity for comparative studies

    • Assessment of potential cross-reactivity with different APPL2 isoforms

  • Validation Planning Tools:

    • Automated design of validation experiments based on antibody characteristics

    • Statistical power calculators for designing validation studies

    • Simulation of experimental outcomes under different conditions

    • Optimization of antibody concentration and conditions for specific applications

These computational approaches can significantly reduce the time and resources required for APPL2 antibody development while improving specificity and performance across applications. Integration of computational design with high-throughput experimental validation represents the future of antibody technology for APPL2 and other research targets.

What are the key considerations for designing robust APPL2 antibody-based experiments?

Designing robust APPL2 antibody-based experiments requires careful attention to multiple factors:

  • Experimental Design Fundamentals:

    • Begin with clear research questions about APPL2 function or expression

    • Include appropriate positive and negative controls

    • Plan for biological and technical replicates

    • Consider power analysis to determine sample size requirements

    • Design experiments to test alternative hypotheses

  • Antibody Selection and Validation:

    • Choose antibodies validated for your specific application (WB, IHC, etc.)

    • Verify specificity through knockout/knockdown controls when possible

    • Consider using multiple antibodies targeting different APPL2 epitopes

    • Validate cross-reactivity if working with non-human species

    • Document antibody characteristics including lot number, concentration, and source

  • Technical Optimization:

    • Determine optimal antibody concentration through titration experiments

    • Optimize antigen retrieval methods for fixed tissues

    • Establish appropriate blocking conditions to minimize background

    • Standardize incubation times and temperatures

    • Develop consistent quantification methods

  • Data Interpretation:

    • Establish clear criteria for positive/negative results before analysis

    • Use quantitative methods when possible rather than subjective assessments

    • Consider biological context when interpreting APPL2 expression patterns

    • Be aware of potential artifacts and technical limitations

    • Triangulate findings with orthogonal techniques

  • Reporting Standards:

    • Document detailed methods following ARRIVE or similar guidelines

    • Report antibody validation data and negative results

    • Provide quantitative data with appropriate statistical analysis

    • Include representative images showing the range of results observed

    • Share detailed protocols to enhance reproducibility

By addressing these considerations systematically, researchers can generate more reliable and reproducible data about APPL2 expression and function, advancing understanding of this important protein's roles in normal physiology and disease states.

What emerging research areas might benefit from improved APPL2 antibody techniques?

Several emerging research areas stand to benefit significantly from advances in APPL2 antibody technology:

  • Metabolic Disease Research:

    • Given APPL2's role in carbohydrate metabolism and homeostasis

    • Investigation of APPL2 expression in diabetes and obesity models

    • Study of APPL2's interactions with insulin signaling pathways

    • Examination of APPL2 modifications in response to metabolic stress

  • Neurodegenerative Disease Studies:

    • Leveraging APPL2's notable expression in brain tissue

    • Analysis of APPL2 distribution in specific neuronal populations

    • Investigation of potential roles in neuronal survival and function

    • Examination of APPL2 alterations in Alzheimer's and Parkinson's disease models

  • Cancer Biology:

    • Exploring APPL2's involvement in cell cycle regulation

    • Comparison of APPL2 expression in normal versus neoplastic tissues

    • Investigation of APPL2's role in cancer cell metabolism

    • Analysis of APPL2-dependent signaling pathways in tumor progression

  • Developmental Biology:

    • Studying APPL2 expression patterns during embryonic development

    • Investigation of APPL2's role in tissue differentiation

    • Examination of APPL2 function in stem cell biology

    • Cross-species comparison of APPL2 expression in evolutionarily conserved developmental pathways

  • Systems Biology Integration:

    • Mapping APPL2 protein interaction networks across cell types

    • Integration of APPL2 expression data with transcriptomic and proteomic datasets

    • Development of computational models of APPL2-dependent pathways

    • Investigation of APPL2's role in cellular responses to environmental stimuli

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.