ALPL Antibody

Shipped with Ice Packs
In Stock

Description

Introduction

The Alkaline Phosphatase, Liver/Bone/Kidney (ALPL) antibody is a specialized immunological reagent designed to detect and analyze the ALPL enzyme, a key biomarker involved in bone mineralization, liver function, and cellular differentiation. This antibody has become a critical tool in both basic research and clinical diagnostics, particularly in studying metabolic bone disorders, liver diseases, and certain cancers. Below is a detailed analysis of its structure, applications, product specifications, and research findings, supported by diverse scientific sources.

Structure and Types of ALPL Antibodies

ALPL antibodies are categorized into polyclonal and monoclonal types, differing in their production methods and specificity:

  • Polyclonal Antibodies: Produced in rabbits or mice by immunizing with recombinant ALPL fusion proteins (e.g., amino acids 230–440 of human ALPL) . These antibodies recognize multiple epitopes, offering broader reactivity but lower specificity.

  • Monoclonal Antibodies: Generated via hybridoma technology, such as the TRA-2-49/6E clone (mouse IgG1) , which targets the ALPL isoenzyme with high specificity.

ProductTypeHostReactivityImmunogen
CAB12396PolyclonalRabbitHuman, Mouse, RatRecombinant ALPL (aa 230–440)
AF2910MonoclonalMouseMouse, RatNative ALPL (embryonic stem cells)
TRA-2-49/6EMonoclonalMouseHuman, PrimateEmbryonal carcinoma cells (EC 2102Ep)

3.1. Research Applications

  • Western Blot (WB): Detects ALPL protein in lysates (e.g., HeLa cells, osteosarcoma cell lines) .

  • Immunohistochemistry (IHC): Localizes ALPL in tissues, such as liver bile canaliculi or osteosarcoma metastases .

  • Immunocytochemistry (ICC): Stains ALPL on cell surfaces (e.g., rat mesenchymal stem cells) .

  • Chimeric Antigen Receptor (CAR) T-Cell Therapy: Targets ALPL-1 isoforms in osteosarcoma (OS), showing preclinical efficacy .

3.2. Diagnostic Applications

  • ELISA: Measures ALPL levels in serum or conditioned media .

  • Flow Cytometry: Identifies ALPL-expressing cells (e.g., OS patient-derived xenografts) .

4.1. Bone Metabolism and Osteosarcoma

ALPL is overexpressed in osteosarcoma (OS), making it a therapeutic target. CAR-T cells engineered to recognize ALPL-1 isoforms exhibit robust antitumor activity in preclinical OS models without off-target effects on healthy tissues .

4.2. Liver and Kidney Pathologies

  • Elevated ALPL levels correlate with liver damage (e.g., hepatitis, cirrhosis) .

  • Renal tubular ALPL expression is linked to phosphate metabolism disorders .

4.3. Developmental Biology

ALPL is critical during embryogenesis, particularly in bone mineralization and stem cell differentiation. Antibody-based studies reveal its role in regulating osteoblast activity .

5.1. Key Products

ProductHostReactivityApplicationsCitations
CAB12396RabbitHuman, Mouse, RatWB, IHC, ELISA
AF2910MouseMouse, RatFlow Cytometry, WB
TRA-2-49/6EMouseHuman, PrimateIHC, WB
11187-1-APRabbitHumanWB, IHC, ICC, ELISA

5.2. Immunogen Sequences

  • CAB12396: Recombinant ALPL (aa 230–440) .

  • TRA-2-49/6E: Full-length ALPL from embryonal carcinoma cells .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Generally, we can ship your order within 1-3 business days after receiving it. The delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery information.
Synonyms
AKP2 antibody; Alkaline phosphatase liver/bone/kidney antibody; Alkaline phosphatase liver/bone/kidney isozyme antibody; Alkaline phosphatase tissue nonspecific isozyme antibody; Alkaline phosphatase, tissue-nonspecific isozyme antibody; Alkaline phosphomonoesterase antibody; Alpl antibody; AP TNAP antibody; AP-TNAP antibody; APTNAP antibody; BAP antibody; FLJ40094 antibody; FLJ93059 antibody; Glycerophosphatase antibody; HOPS antibody; Liver/bone/kidney type alkaline phosphatase antibody; MGC161443 antibody; MGC167935 antibody; PHOA antibody; PPBT_HUMAN antibody; Tissue non specific alkaline phosphatase antibody; Tissue nonspecific ALP antibody; TNAP antibody; TNSALP antibody
Target Names
Uniprot No.

Target Background

Function
Alkaline phosphatase (ALPL) is an enzyme that plays a crucial role in various biological processes, including skeletal mineralization, adaptive thermogenesis, and neurotransmitter synthesis. ALPL metabolizes diverse phosphate compounds, displaying broad substrate specificity. While it can hydrolyze a wide range of compounds, only a few, such as diphosphate (inorganic pyrophosphate; PPi), pyridoxal 5'-phosphate (PLP), and N-phosphocreatine, are considered natural substrates.

In skeletal mineralization, ALPL is essential for the proper formation of bones and teeth. It hydrolyzes extracellular diphosphate, a known inhibitor of mineralization, into phosphate. This action promotes the formation of hydroxyapatite crystals and increases the concentration of inorganic phosphate. ALPL works in conjunction with PHOSPHO1, another enzyme involved in mineralization, but with a distinct role. While PHOSPHO1 initiates hydroxyapatite crystallization in matrix vesicles, ALPL catalyzes the expansion of hydroxyapatite crystallization within the extracellular matrix.

ALPL also contributes to skeletal mineralization by dephosphorylating osteopontin (SSP1), a protein that inhibits hydroxyapatite crystallization in its phosphorylated state. However, the exact mechanism of SSP1 dephosphorylation by ALPL, whether direct or indirect, remains unclear.

Regarding neurotransmitter synthesis, ALPL catalyzes the dephosphorylation of PLP to pyridoxal (PL), the transportable form of vitamin B6. This process ensures sufficient PLP availability in the brain, which is vital as a cofactor for enzymes involved in the synthesis of various neurotransmitters.

Beyond its role in skeletal mineralization and neurotransmitter synthesis, ALPL also regulates the availability of ligands for purinergic receptors by mediating the stepwise degradation of ATP to adenosine. Additionally, it dephosphorylates microbial products, such as lipopolysaccharides (LPS), and other phosphorylated small molecules, like poly-inosine:cytosine (poly I:C).

In the context of adaptive thermogenesis, ALPL functions as a key regulator. It localizes to the mitochondria of thermogenic fat cells and facilitates the hydrolysis of N-phosphocreatine, initiating a futile cycle of creatine dephosphorylation and phosphorylation. This futile creatine cycle dissipates the high energy charge of N-phosphocreatine as heat without performing any mechanical or chemical work, thus contributing to thermogenesis.
Gene References Into Functions
  1. A significant proportion of adult heterozygotes for ALPL mutations may exhibit nonspecific symptoms not directly attributable to their heterozygosity. PMID: 29236161
  2. The ALPL SNP, rs1256328, was identified as a significant association with kidney stone disease status in a large Chinese Han cohort. PMID: 29489416
  3. The expression of ALP mRNA and ALP activity in bone tissue was significantly higher in osteoporosis patients with fractures compared to those without fractures. PMID: 29786747
  4. Genu varum is associated with alkaline phosphatase levels, irrespective of the presence of radiographic abnormalities in the growth plate in children. PMID: 28664247
  5. Adults with persistent hypophosphatasemia frequently harbor alkaline phosphatase mutations and exhibit elevated ALP substrates. PMID: 28401263
  6. Mutations in ALPL leading to reduced alkaline phosphatase activity are responsible for Hypophosphatasia, a rare disorder characterized by defective bone and teeth mineralization and premature tooth loss. PMID: 28570402
  7. Research indicates that increased TNAP activity in ACDC (arterial calcification due to deficiency of CD73) contributes to ectopic calcification by disrupting the extracellular balance of PPi and Pi. This suggests potential therapeutic targets for ACDC. PMID: 27965423
  8. The identification of 11 novel ALPL mutations in five different HPP forms and the observation of a recurrent mutation, p. (Thr166Ile) in the Spanish population, expand our understanding of pathogenic ALPL mutations. PMID: 28127875
  9. Preoperative calcitonin levels were correlated with the presence of tumor, whereas alkaline phosphatase (ALP) levels were not. Tumor volume was not significantly associated with ALP or calcitonin levels in the preoperative or postoperative periods. During long-term follow-up, serum ALP was significantly associated with tumor recurrence, but serum calcitonin was not. PMID: 27922893
  10. A patient's ALPL gene mutation was traced back to a c.228delG mutation inherited from his mother and a c.407G>A compound heterozygous mutation from his father. PMID: 28506345
  11. Both PPARgamma gene expression and TNALP activity increased during intracellular lipid accumulation in HepG2 and 3T3-L1 cells. Inhibition of TNALP blocked intracellular lipid accumulation but did not alter the expression of the PPARgamma gene. PMID: 28209522
  12. ALPL is a major contributor to the pathogenesis of prostate cancer progression. PMID: 28006818
  13. Results indicated that the 1559delT mutant was not retained on the plasma membrane due to a lack of the Glycosylphosphatidylinositol anchor. PMID: 27680481
  14. ALPL expression is significantly upregulated in human masticatory mucosa during wound healing. PMID: 28005267
  15. Serum ALP levels were not associated with increased death risk in prevalent HD patients over a 5-year interval. PMID: 27467278
  16. In patients with bone metastases, combined detection of BSP, ALP, ICTP, and PSA markers could improve the positive-predictive value. PMID: 27323113
  17. Studies reveal that amino acid substitutions at position 426 of TNSALP differentially affect the structure and function of TNSALP, providing insights into the molecular and cellular basis of hypophosphatasia. PMID: 28000043
  18. Half of adult individuals with unexplained low serum ALP carried an ALPL mutation. The presence of a mutated allele was associated with tooth loss, slightly lower serum ALP levels, higher levels of pyridoxal phosphate and phosphoethanolamine, and mildly increased serum phosphate. PMID: 26783040
  19. Dynamic changes in ALP, LDH, and PSA during Abiraterone therapy are associated with optimal clinical benefit and overall survival (OS) in bone metastatic castration-resistant prostate cancer. PMID: 26975660
  20. Glycosylation differences in human bone alkaline phosphatases are crucial for protein-protein interactions with collagen type I. PMID: 26645431
  21. Analysis of a series of multiple N-glycan depletion mutants in TNSALP revealed that three N-glycans on N230, N271, and N303 were the minimal requirement for the structure and function of TNSALP and a prerequisite for its stable expression in a cell. PMID: 26797772
  22. The presence of TNAP increased the dynamics and decreased the ordering of model membranes. PMID: 26389140
  23. TNAP is co-expressed by dental pulp stromal cells alongside other bone marrow stromal cell markers, and cell density influences TNAP expression levels. PMID: 25636587
  24. ALP quartiles were significantly associated with albuminuria in participants with estimated glomerular filtration rate >120. Higher ALP levels are significantly associated with renal hyperfiltration. PMID: 25853240
  25. During skeletal mineralization, the building Ca2+ gradient initially activates TNAP but gradually inactivates it at high Ca2+ concentrations as mineralization nears completion. PMID: 25775211
  26. TNAP activity is significantly increased in the brain in both sporadic and familial forms of Alzheimer's Disease (AD) and in the plasma of AD patients. PMID: 26219720
  27. A non-linear relationship exists between serum levels of ALP and phosphate and the risk of total mortality from cardiovascular diseases. PMID: 25033287
  28. Higher alkaline phosphatase was associated with short-term adverse outcomes of peritoneal dialysis-related peritonitis. PMID: 25246707
  29. ALP mRNA binds to and is stabilized by vimentin. PMID: 25536665
  30. Elevated AP was associated with the presence of COPD and respiratory symptoms (cough, wheezing). PMID: 25336462
  31. A two-month alkaline phosphatase level of <100 U/L exhibited a negative predictive value of 97% for the development of ischemic cholangiopathy after liver transplantation. PMID: 25769592
  32. A novel role for alkaline phosphatase in ERK1 and ERK2 dephosphorylation has been identified in renal cell carcinoma cell lines. PMID: 25241253
  33. In inflammatory cholestatic conditions, a loss of liver AP activity may promote hyper-adenosine triphosphate-bilia, lipopolysaccharide overload, and subsequent exacerbation and perpetuation of inflammation. [review] PMID: 25603770
  34. Patterns were confirmed in human teeth, including widespread TNAP and NPP1 restricted to cementoblasts lining acellular cementum. PMID: 25504209
  35. High levels of alkaline phosphatase (a biochemical marker of osteosynthesis) are associated with a poor prognosis in metastatic bone cancer originating from disseminated breast cancer. PMID: 25342482
  36. Polymorphisms in ALP, ENPP1, and ANKH are significant genetic risk factors contributing to Pseudoxanthoma elasticum. PMID: 25025693
  37. REVIEW: role of bone-type tissue-nonspecific alkaline phosphatase and PHOSPO1 in vascular calcification PMID: 24533943
  38. This family report indicates that mapping ALPL mutations within the gene does not necessarily predict the clinical severity of the hypophosphatasia phenotype. PMID: 24569605
  39. Data suggest that alkaline phosphatase (AP) velocity kinetics (APV) is an independent predictor of overall survival (OS) and bone metastasis-free survival (BMFS) in patients with castration-resistant prostate cancer (CRPC). PMID: 24929891
  40. This research explores the novel role of ALP in cell viability and apoptosis and its involvement in renal cell carcinoma tumorigenesis. PMID: 24909115
  41. Effect of cyclic mechanical stimulation on the expression of osteogenesis genes in human intraoral mesenchymal stromal and progenitor cells. PMID: 24804200
  42. DNMT inhibitors facilitate the Pi-induced development of vascular calcification through the upregulation of ALP expression, accompanied by a reduction in the DNA methylation level of the ALP promoter region. PMID: 24441913
  43. The ABO locus is a major determinant for serum ALP levels in the Chinese Han population. PMID: 24094242
  44. The CPT score, alkaline phosphatase > 1.5 ULN, and the CS nonresponse had an independent impact on 90-day survival in alcoholic hepatitis. PMID: 24151614
  45. This study characterized a novel genetic alteration (c.1318_1320delAAC, p.N440del) in the ALPL gene resulting in odonto-hypophosphatasia (HPP) in monozygotic twins. These findings help define genotype-phenotype associations for odonto-HPP and identify the collagen-binding site as a region of potential structural importance for TNAP function in biomineralization. PMID: 23791648
  46. Serum ALP is adversely associated with measures of arterial structure and function in hypertensive African men. PMID: 22656046
  47. The Y28D, A111T, and T389N mutants displayed negligible ALP activity in vitro compared to the wild-type (WT) tissue-nonspecific alkaline phosphatase. PMID: 24022022
  48. The most frequent clinical type was the PLH type, with prognosis related to respiratory failure, biochemical/radiological changes, and ALPL mutations. PMID: 24276437
  49. This study investigated two mineralization-related genes, TNAP and ANKH polymorphisms associated with ankylosing spondylitis (AS) in the North Chinese Han population. PMID: 23612078
  50. Data suggest that the promineralization role of TNAP may be related not only to its known pyrophosphatase activity but also to its ability to modify the phosphorylation status of OPN. PMID: 23427088

Show More

Hide All

Database Links

HGNC: 438

OMIM: 146300

KEGG: hsa:249

STRING: 9606.ENSP00000363965

UniGene: Hs.75431

Involvement In Disease
Hypophosphatasia (HOPS); Hypophosphatasia childhood type (HOPSC); Hypophosphatasia infantile type (HOPSI)
Protein Families
Alkaline phosphatase family
Subcellular Location
Cell membrane; Lipid-anchor, GPI-anchor. Extracellular vesicle membrane; Lipid-anchor, GPI-anchor. Mitochondrion membrane; Lipid-anchor, GPI-anchor. Mitochondrion intermembrane space.

Q&A

What is ALPL and why is it important in research?

ALPL (Alkaline Phosphatase, Liver/Bone/Kidney isozyme) is a critical enzyme involved in bone mineralization and plays key roles in regulating various cellular processes. This enzyme functions by removing phosphate groups from molecules such as DNA, RNA, and proteins at high pH conditions. ALPL is particularly significant in research because its dysregulation has been implicated in multiple pathological conditions including metabolic bone disorders, liver diseases, and certain cancers . The enzyme is expressed at high concentrations in tissues such as liver, bile ducts, placenta, and bone, making it an important marker for studies in these areas . Understanding ALPL expression patterns and activity levels provides valuable insights into both normal physiological processes and disease mechanisms.

What are the different types of ALPL antibodies available for research?

Researchers can utilize several types of ALPL antibodies depending on their specific experimental needs:

  • Monoclonal vs. Polyclonal: Polyclonal antibodies like CAB12396 recognize multiple epitopes of ALPL and are produced in rabbits, offering broad detection capabilities . Monoclonal antibodies like those referenced in source provide higher specificity for particular epitopes, enabling more targeted analyses.

  • Species-specific reactivity: Most commercial ALPL antibodies demonstrate reactivity against human, mouse, and rat ALPL, with varying degrees of cross-reactivity . This is particularly important when designing multi-species studies or translational research.

  • Application-optimized antibodies: Different antibodies are validated for specific applications:

    • Western blot-optimized antibodies (including Picoband® antibodies for high sensitivity)

    • IHC-optimized antibodies for tissue localization studies

    • Flow cytometry-validated antibodies for cellular analyses

When selecting an ALPL antibody, researchers should carefully evaluate the validation data for their specific application and tissue/cell type to ensure optimal experimental outcomes.

How is the specificity of ALPL antibodies validated?

Validation of ALPL antibody specificity typically follows multiple complementary approaches:

  • CRISPR/Cas9 knockout validation: As demonstrated in source , CRISPR-mediated knockout of the ALPL gene in cell lines provides definitive evidence of antibody specificity when staining is abolished in knockout cells compared to wild-type cells.

  • Cross-platform confirmation: Reliable ALPL antibodies show consistent staining patterns across multiple detection platforms (e.g., Western blot, IHC, and IF) . Comparison of staining patterns using different antibodies against the same target (as seen with HPA008765 and HPA007105 antibodies) provides additional validation .

  • Molecular weight verification: Western blot analysis should detect ALPL at its expected molecular weight of approximately 57-80 kDa (the observed 80 kDa band likely reflects glycosylation of the 57 kDa core protein) .

  • Cross-reactivity testing: Comprehensive validation includes testing against non-target tissues and cell lines to confirm minimal background and non-specific binding .

Such multi-parameter validation approaches ensure that research findings based on ALPL antibody staining accurately reflect true biological phenomena rather than technical artifacts.

What are the optimal conditions for using ALPL antibodies in Western blot applications?

For optimal Western blot detection of ALPL using antibodies, researchers should follow these methodological guidelines:

  • Sample preparation:

    • Load 30 μg of protein per lane under reducing conditions

    • Effective lysis buffers should contain phosphatase inhibitors to preserve ALPL's native state

  • Electrophoresis parameters:

    • Use 5-20% gradient SDS-PAGE gels for optimal separation

    • Run at 70V (stacking)/90V (resolving) for 2-3 hours

  • Transfer conditions:

    • Transfer to nitrocellulose membrane at 150 mA for 50-90 minutes

    • Verify transfer efficiency with reversible protein staining

  • Antibody dilutions and incubation:

    • Block with 5% non-fat milk in TBS for 1.5 hours at room temperature

    • Use primary antibody at 1:5000 dilution overnight at 4°C

    • Wash with TBS-0.1% Tween (3 × 5 minutes)

    • Incubate with HRP-conjugated secondary antibody at 1:5000 for 1.5 hours at room temperature

  • Detection optimization:

    • Develop using enhanced chemiluminescence (ECL) detection methods

    • Expected band size is approximately 80 kDa, though the calculated molecular weight is 57 kDa due to post-translational modifications

Researchers should note that ALPL expression varies significantly between cell lines, with high expression observed in liver-derived cells (HepG2) and lower expression in some cancer cell lines like Jurkat .

How should ALPL antibodies be used for immunohistochemistry and immunofluorescence studies?

For successful immunohistochemistry (IHC) and immunofluorescence (IF) studies using ALPL antibodies:

  • Tissue preparation for IHC:

    • Formalin-fixed, paraffin-embedded (FFPE) tissues should be sectioned at 4-6 μm thickness

    • Perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) for optimal antigen retrieval

    • Include appropriate positive control tissues such as liver, which shows strong ALPL expression

  • Cell preparation for IF:

    • Fix cells with 4% paraformaldehyde for 15 minutes at room temperature

    • Permeabilize with 0.1% Triton X-100 in PBS if targeting intracellular epitopes

    • Block with 1-5% BSA or normal serum from the same species as the secondary antibody

  • Antibody incubation parameters:

    • Dilute primary antibodies appropriately (typically 1:100-1:500 for IHC/IF applications)

    • Incubate overnight at 4°C for maximum sensitivity

    • Include negative controls (omitting primary antibody or using isotype controls)

  • Signal detection considerations:

    • For IHC: Use DAB (3,3'-diaminobenzidine) or AEC (3-amino-9-ethylcarbazole) chromogens

    • For IF: Select fluorophores with minimal spectral overlap if performing multiplexing

    • Counterstain nuclei with DAPI or hematoxylin as appropriate

  • Interpretation guidelines:

    • ALPL typically shows membranous and cytoplasmic localization

    • Expression patterns vary by tissue type, with particularly strong staining in liver, bone, and kidney tissues

What are the recommended protocols for flow cytometry using ALPL antibodies?

For flow cytometric detection of ALPL using specific antibodies, researchers should follow these methodological steps:

  • Cell preparation:

    • Harvest cells using enzyme-free cell dissociation solutions to preserve surface epitopes

    • Wash cells in PBS/2% FBS buffer (flow buffer)

    • Adjust cell concentration to 1 × 10^6 cells/100 μl for staining

  • Antibody staining procedure:

    • For surface ALPL: Incubate live cells with primary ALPL antibody (diluted in flow buffer) for 30-60 minutes on ice

    • For intracellular ALPL: Fix cells with 4% paraformaldehyde, permeabilize with 0.1% saponin or commercial permeabilization buffers, then stain

    • Wash twice with flow buffer after primary antibody incubation

  • Secondary antibody considerations:

    • If using unconjugated primary antibodies, incubate with fluorophore-conjugated secondary antibodies for 30 minutes on ice

    • Include appropriate compensation controls when using multiple fluorophores

  • Controls and validation:

    • Include an isotype control antibody at matching concentration

    • Use ALPL-knockout cells (e.g., CRISPR-generated) as negative controls

    • Include known positive cell lines (e.g., osteosarcoma cell lines) as positive controls

  • Analysis parameters:

    • Gate on viable cells using appropriate viability dyes

    • Analyze ALPL expression as mean fluorescence intensity (MFI) relative to isotype control

    • For quantitative analysis of antigen density, use antibody-binding capacity (ABC) beads for calibration

As demonstrated in research applications, this approach can effectively distinguish ALPL-positive populations and evaluate expression levels across different cell types, including tumor cells and normal tissue controls .

How can researchers address non-specific binding issues when using ALPL antibodies?

Non-specific binding is a common challenge when working with ALPL antibodies. To minimize these issues, researchers should implement these methodological solutions:

  • Optimization of blocking conditions:

    • Extend blocking time to 2 hours at room temperature using 5% non-fat milk or BSA

    • Consider alternative blocking agents such as normal serum (from the same species as the secondary antibody)

    • For tissues with high endogenous biotin, use avidin/biotin blocking kits before antibody application

  • Antibody dilution optimization:

    • Perform titration experiments to determine optimal antibody concentration

    • High-quality polyclonal antibodies like Picoband® are specifically designed to provide strong signals with minimal background in Western blot applications

    • Generally, more dilute antibody solutions (1:5000-1:10000 for Western blot) can reduce non-specific binding while maintaining specific signal

  • Additional washing steps:

    • Increase the number and duration of washes (e.g., 5 × 10 minutes)

    • Use detergent-containing wash buffers (0.1-0.3% Tween 20 or Triton X-100) to reduce hydrophobic interactions

  • Pre-adsorption protocols:

    • If cross-reactivity with related proteins is suspected, pre-incubate the antibody with recombinant related proteins

    • For polyclonal antibodies, consider using purified antigen for pre-adsorption to improve specificity

  • Negative control validation:

    • Include ALPL-knockout cells or tissues as negative controls to definitively identify non-specific signals

    • As demonstrated in CRISPR/Cas9 knockout studies, specific ALPL antibodies should show no binding to ALPL-knockout samples

These approaches systematically address the most common sources of non-specific binding when working with ALPL antibodies, leading to cleaner, more interpretable experimental results.

What factors affect the detection of ALPL in different tissue types?

Detection of ALPL across diverse tissue types presents several challenges due to varying expression levels and tissue-specific characteristics. Key factors that influence detection include:

  • Tissue-specific expression patterns:

    • ALPL is highly expressed in liver, bone, kidney, and placenta, making these tissues ideal positive controls

    • Expression can vary dramatically between tissue types, necessitating optimization of antibody concentration for each tissue

    • Immunohistochemical studies have demonstrated distinct staining patterns in human adrenal gland, endometrium, liver, and pancreas

  • Fixation and processing effects:

    • Overfixation can mask epitopes, particularly for membrane-bound proteins like ALPL

    • Optimize fixation time (8-24 hours in 10% neutral buffered formalin) for most consistent results

    • Antigen retrieval methods must be tissue-specifically optimized (e.g., heat-induced vs. enzymatic retrieval)

  • Endogenous enzyme activity interference:

    • Tissues with high endogenous phosphatase activity can create background issues

    • Use levamisole (5 mM) to inhibit endogenous alkaline phosphatase when using alkaline phosphatase-based detection systems

    • For immunohistochemistry applications, peroxidase-based detection may be preferable to avoid this interference

  • Sample handling and storage conditions:

    • ALPL enzyme activity is temperature-sensitive, which can affect epitope preservation

    • Flash-frozen samples may preserve epitopes better than FFPE tissues for certain antibody clones

    • Archived samples may show reduced immunoreactivity due to epitope degradation over time

  • Multi-antibody validation approach:

    • As demonstrated in source , comparing staining patterns of independent antibodies targeting different ALPL epitopes (HPA008765 and HPA007105) across tissues provides more reliable results and confirms genuine expression patterns

Understanding these factors allows researchers to develop tissue-specific protocols that maximize detection sensitivity and specificity.

How do post-translational modifications affect ALPL antibody binding?

Post-translational modifications (PTMs) significantly impact ALPL antibody binding and can affect experimental outcomes. Researchers should consider these methodological implications:

Understanding these PTM effects is essential for accurate interpretation of ALPL antibody-based experimental results, particularly when comparing different physiological or pathological states.

How can ALPL antibodies be utilized in CAR-T cell therapy development?

ALPL antibodies are emerging as crucial tools in developing chimeric antigen receptor (CAR) T cell therapies, particularly for osteosarcoma. Based on recent research, here's a methodological framework for utilizing ALPL antibodies in this advanced application:

  • Identification of ALPL as a therapeutic target:

    • ALPL-1 has been identified as the target of TP-1 and TP-3 antibodies, which show specificity for osteosarcoma with limited cross-reactivity to normal tissues

    • Researchers can use commercial ALPL antibodies to validate expression patterns across tumor samples and normal tissues to assess target specificity

  • Antibody-based CAR design methodology:

    • Extract coding sequences from hybridomas producing anti-ALPL antibodies (e.g., TP-1, TP-3)

    • Engineer these sequences into CAR constructs (e.g., OSCAR-1, OSCAR-3) for T cell redirection against ALPL-expressing tumors

    • Validate CAR construct expression and functionality using flow cytometry with anti-ALPL antibodies

  • Target validation using CRISPR/Cas9:

    • Generate ALPL knockout cell lines using CRISPR/Cas9 technology

    • Confirm knockout efficiency using commercial anti-ALPL antibodies via flow cytometry and Western blot

    • Demonstrate absence of CAR-T cell activity against knockout cells to confirm specificity

  • Preclinical efficacy and safety assessment:

    • Evaluate CAR-T cell cytotoxicity against ALPL-expressing tumor cells using bioluminescence-based killing assays

    • Assess potential off-tumor toxicity by testing CAR-T cell activity against normal cells/tissues with low ALPL expression

    • Use immunohistochemistry with anti-ALPL antibodies to screen for expression in normal tissues to predict potential toxicities

  • ALPL surface density quantification:

    • Utilize flow cytometry with calibrated anti-ALPL antibodies to determine antigen density on tumor cells

    • Compare antigen density between tumor and normal cells to establish therapeutic window

    • This quantitative approach helps predict CAR-T efficacy and potential for on-target, off-tumor toxicity

This advanced application demonstrates how ALPL antibodies extend beyond basic research tools to therapeutic development platforms, highlighting their potential in precision medicine approaches for ALPL-expressing malignancies.

What are the methodological considerations for studying ALPL in bone metabolism disorders?

Investigating ALPL in bone metabolism disorders requires specialized methodological approaches due to its critical role in bone mineralization. Researchers should consider these advanced technical strategies:

  • Tissue-specific analysis protocols:

    • For bone samples: Decalcification procedures must be carefully optimized as they can affect ALPL epitope preservation

    • Use EDTA-based slow decalcification (rather than acid-based rapid methods) to better preserve ALPL antigenicity

    • Consider undecalcified plastic-embedded sections with specialized cutting techniques for optimal preservation of ALPL in its native microenvironment

  • Distinguishing ALPL isoforms:

    • ALPL exists in multiple isozymes including liver/bone/kidney (tissue non-specific), intestinal, and placental forms

    • Select antibodies that can specifically distinguish the bone ALPL isoform from other isozymes

    • Validate isoform specificity using cells expressing single isozymes as controls

  • Correlation of expression with enzyme activity:

    • Combine immunodetection of ALPL protein (using specific antibodies) with enzymatic activity assays

    • Use histochemical alkaline phosphatase activity staining on serial sections to correlate protein levels with functional activity

    • This dual approach distinguishes changes in expression versus changes in enzymatic activity in pathological conditions

  • Osteoblast differentiation studies:

    • Track ALPL expression during osteoblast differentiation using time-course immunofluorescence or flow cytometry

    • Compare ALPL antibody staining with osteogenic markers to establish temporal relationships during differentiation

    • Use mesenchymal stem cells as negative controls when assessing osteoblast ALPL expression patterns

  • Advanced multiplexing approaches:

    • Implement multiplex immunofluorescence to simultaneously visualize ALPL and other bone metabolism markers

    • This approach can reveal co-expression patterns and spatial relationships between ALPL and regulatory factors

    • Combine with image analysis software for quantitative assessment of expression levels across different pathological states

These methodological considerations enable researchers to conduct sophisticated investigations into ALPL's role in bone disorders, potentially revealing novel therapeutic targets for conditions like hypophosphatasia and other metabolic bone diseases.

How can researchers integrate ALPL antibody data with -omics approaches?

Integrating ALPL antibody-derived data with multi-omics approaches creates powerful research paradigms for understanding ALPL biology in complex systems. Here's a methodological framework for this advanced research application:

  • Correlative proteogenomic analysis:

    • Compare ALPL protein levels (detected by antibodies via Western blot or IHC) with mRNA expression (from RNA-seq)

    • Identify post-transcriptional regulatory mechanisms when protein and mRNA levels are discordant

    • This approach has revealed elevated ALPL mRNA in osteosarcoma samples that corresponds with protein detection using antibodies, validating ALPL as a potential therapeutic target

  • Antibody-based ChIP-seq methodology:

    • Use ALPL antibodies to identify protein interaction partners through chromatin immunoprecipitation followed by sequencing (ChIP-seq)

    • This approach can reveal transcriptional networks associated with ALPL expression and function

    • Validate interactions using reciprocal co-immunoprecipitation with antibodies against predicted partners

  • Spatial transcriptomics integration:

    • Combine antibody-based ALPL protein localization (via immunofluorescence) with spatial transcriptomics data

    • Map protein expression patterns to transcriptional signatures in specific tissue microenvironments

    • This integrated approach provides insights into how ALPL expression relates to local cellular functions and signaling networks

  • Single-cell multi-omics correlation:

    • Use flow cytometry with ALPL antibodies to isolate ALPL-positive cell populations

    • Perform single-cell RNA-seq, ATAC-seq, or proteomics on isolated populations

    • Compare molecular profiles of ALPL-high versus ALPL-low cells to identify regulatory mechanisms and downstream pathways

  • Quantitative antibody-based assays for clinical correlation:

    • Develop quantitative ALPL antibody-based assays (ELISA, multiplex bead arrays) for patient samples

    • Correlate protein levels with clinical data, genomic alterations, and treatment outcomes

    • Identify biomarker potential of ALPL for patient stratification in personalized medicine approaches

This integrated approach transforms ALPL antibody-based detection from a simple protein identification tool to a sophisticated component of multi-dimensional biological analysis, providing deeper insights into ALPL's role in health and disease.

What methodological approaches are recommended for studying ALPL in cancer research?

Cancer research involving ALPL requires specific methodological considerations due to its variable expression across tumor types and potential as a therapeutic target. Researchers should implement these specialized approaches:

  • Patient-derived xenograft (PDX) model development:

    • Establish PDX models from primary tumors expressing ALPL (especially osteosarcoma)

    • Validate ALPL expression in PDX models using antibody-based flow cytometry and IHC

    • Research indicates PDX models maintain high ALPL expression levels comparable to patient samples, making them valuable for therapeutic testing

  • Tumor microenvironment analysis:

    • Employ multiplex immunofluorescence with ALPL antibodies combined with markers for immune cells, stromal components, and other tumor microenvironment elements

    • This approach reveals spatial relationships between ALPL-expressing tumor cells and their microenvironment

    • Quantify ALPL expression gradients relative to vascular markers or hypoxic regions to understand its regulation in tumor contexts

  • Circulating tumor cell detection:

    • Develop flow cytometry protocols using anti-ALPL antibodies to identify and isolate circulating tumor cells

    • Combine with epithelial markers (CK, EpCAM) and exclude hematopoietic markers (CD45)

    • This methodology provides insights into metastatic potential of ALPL-expressing tumors

  • Therapeutic response monitoring:

    • Use ALPL antibodies to monitor expression changes before and after treatment

    • Deploy quantitative image analysis of IHC or IF staining to measure therapy-induced changes in ALPL expression

    • Correlate expression patterns with treatment outcomes to identify predictive biomarker potential

  • ALPL-directed therapeutic development pipeline:

    • Screen patient samples using ALPL antibodies to identify high-expressing tumors

    • Develop ALPL-targeted therapies including antibody-drug conjugates or CAR-T cells

    • Monitor on-target, off-tumor effects by systematically screening normal tissues for ALPL expression

Tumor TypeALPL Expression (Flow Cytometry)Therapeutic PotentialDetection Antibody
Osteosarcoma cell linesStrong positive (except U2OS)High target potentialTP-3 antibody
Osteosarcoma PDXVery strong positiveExcellent targetTP-3 antibody
Mesenchymal stem cellsNegativeLow off-target concernTP-3 antibody
Normal tissuesComplete negativityFavorable safety profileTP-3 antibody

Table 1: ALPL expression profiles across tumor types and normal tissues, demonstrating potential therapeutic window for ALPL-targeted therapies based on flow cytometry data with TP-3 antibody

These methodological approaches enable systematic investigation of ALPL in cancer contexts, facilitating translation from basic research to therapeutic applications.

How should researchers interpret contradictory ALPL antibody data from different experimental platforms?

Resolving contradictory ALPL antibody data across different experimental platforms requires systematic analytical approaches. Here's a methodological framework for addressing such discrepancies:

  • Cross-platform validation protocol:

    • When conflicting results occur, implement parallel testing using multiple antibodies against different ALPL epitopes

    • Compare results across Western blot, IHC, flow cytometry, and IF to identify platform-dependent variations

    • Research has shown that antibodies like TP-1 and TP-3 can show different staining intensities despite targeting the same antigen, with TP-3 demonstrating higher sensitivity

  • Epitope-specific considerations:

    • Map the specific epitopes recognized by different antibodies (e.g., CAB12396 targets amino acids 230-440 of human ALPL)

    • Epitopes may be differentially accessible depending on protein conformation in different applications

    • Denatured conditions (Western blot) versus native conditions (flow cytometry, IF) can yield different results with the same antibody

  • Antibody validation hierarchy:

    • Establish a validation hierarchy with genetic approaches (CRISPR knockout) as the gold standard

    • As demonstrated in source , CRISPR/Cas9-mediated ALPL knockout provides definitive validation of antibody specificity

    • When knockout validation isn't possible, use multiple antibodies against different epitopes to confirm expression patterns

  • Quantitative data reconciliation approach:

    • Convert semi-quantitative data from different platforms to comparable scales

    • Use positive and negative control samples consistently across all platforms

    • Employ statistical methods to normalize data from different techniques to allow direct comparison

  • Decision matrix for resolving conflicts:

ScenarioPrimary Resolution ApproachSecondary Validation
Western blot ≠ IHCCheck sample preparation, fixation effectsUse fresh frozen samples for both
Flow cytometry ≠ Western blotEvaluate surface vs. total protein expressionSurface biotinylation assay
Antibody A ≠ Antibody BCompare epitopes, validate with knockout controlsCompetitive binding assays
IHC ≠ mRNA expressionConsider post-transcriptional regulationProtein half-life studies

Table 2: Decision matrix for resolving contradictory ALPL data across experimental platforms

This systematic approach transforms contradictory results from a challenge into an opportunity to gain deeper insights into ALPL biology, potentially revealing novel regulatory mechanisms or protein isoforms.

What are the methodological considerations for developing clinical diagnostic assays using ALPL antibodies?

Developing clinical diagnostic assays using ALPL antibodies requires rigorous methodological approaches to ensure reliability, reproducibility, and clinical utility. Researchers should consider these advanced technical requirements:

  • Antibody pair selection and validation:

    • Screen multiple antibody pairs recognizing distinct, non-overlapping epitopes of ALPL

    • Validate antibody performance across diverse sample types (serum, plasma, tissue extracts)

    • Develop sandwich ELISA formats with capture and detection antibody combinations optimized for clinical samples

    • Compare results with established clinical assays measuring ALPL enzymatic activity to ensure correlation

  • Reference standard development:

    • Create recombinant ALPL protein standards with defined concentrations for assay calibration

    • Verify protein standards using mass spectrometry to confirm identity and purity

    • Develop international reference materials to enable cross-laboratory standardization

    • Include controls for known ALPL variants to account for potential epitope variations

  • Assay performance qualification:

    • Determine analytical sensitivity (limit of detection, limit of quantification)

    • Establish assay dynamic range to encompass clinically relevant ALPL concentrations

    • Assess precision (intra-assay and inter-assay coefficients of variation)

    • Evaluate accuracy through spike-recovery and dilution linearity studies

    • Define stability parameters for samples and reagents under various storage conditions

  • Clinical validation strategies:

    • Perform retrospective studies comparing antibody-based ALPL detection with clinical outcomes

    • Establish reference ranges in diverse populations (age, sex, ethnicity)

    • Determine diagnostic sensitivity, specificity, and predictive values for specific clinical applications

    • Compare performance against current gold standard methods (e.g., genetic testing for hypophosphatasia)

  • Technological platform considerations:

PlatformAdvantagesLimitationsKey Development Considerations
ELISAWidely accessible, quantitativeModerate throughputOptimize blocking buffers to minimize matrix effects
Multiplex bead arraysHigh throughput, multiple analytesComplex optimizationCareful cross-reactivity testing with other biomarkers
Point-of-care immunoassaysRapid results, minimal equipmentLower sensitivityAntibody stability at room temperature crucial
Automated clinical analyzersHigh throughput, standardizedPlatform-specific developmentAdaptation to existing clinical workflows

Table 3: Technological platform considerations for ALPL antibody-based clinical assay development

This methodological framework ensures that ALPL antibody-based diagnostics meet the rigorous requirements for clinical implementation, potentially improving diagnostic accuracy for conditions where ALPL levels have clinical significance.

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.