PKD1L2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery time may vary depending on the shipping method and destination. Please consult your local distributor for specific delivery details.
Synonyms
PKD1L2 antibody; KIAA1879 antibody; PC1L2 antibody; Polycystic kidney disease protein 1-like 2 antibody; PC1-like 2 protein antibody; Polycystin-1L2 antibody
Target Names
PKD1L2
Uniprot No.

Target Background

Function
PKD1L2 may function as an ion-channel regulator and may also act as a G-protein-coupled receptor.
Gene References Into Functions
  1. A study in a Korean population identified a 6.2 kb deletion in the PKD1L2 gene associated with colorectal cancer. PMID: 27605020
  2. Genes are mapped to chromosomes, and gene structures are characterized. Protein structure prediction reveals strong ion channel signature motifs, suggesting their potential function as components of cation channel pores. PMID: 12782129
  3. PKD1L2 is expressed in both developing and adult heart and kidney tissue. PMID: 15203210
  4. Ectopic expression of PKD1L2 in transgenic mice resulted in severe muscle atrophy. PMID: 19578180
Database Links

HGNC: 21715

OMIM: 607894

KEGG: hsa:114780

STRING: 9606.ENSP00000469432

UniGene: Hs.413525

Protein Families
Polycystin family
Subcellular Location
Membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in all tissues tested including brain, placenta, mammary gland, testis, lung and liver. Highest expression in skeletal muscle. Isoform 2 is expressed in heart and kidney.

Q&A

What is PKD1L2 and what functional roles has it been implicated in?

PKD1L2 (Polycystic Kidney Disease 1-Like 2) is a member of the polycystin protein family. It contains 11 transmembrane domains, a latrophilin/CL-1-like GPCR proteolytic site (GPS) domain, and a polycystin-1, lipoxygenase, alpha-toxin (PLAT) domain . The protein may function as a component of cation channel pores, an ion-channel regulator, or possibly as a G-protein-coupled receptor .

Research has shown that PKD1L2 is related to PKD1, which is associated with Autosomal Dominant Polycystic Kidney Disease (ADPKD) . Notably, upregulation of PKD1L2 in a mouse model (ostes) has been linked to neuromuscular impairments including neuromuscular junction degeneration, polyneuronal innervation, and myopathy .

What applications are PKD1L2 antibodies commonly used for in research?

PKD1L2 antibodies are utilized in multiple experimental applications:

ApplicationAvailable Antibodies
Western Blotting (WB)Polyclonal and monoclonal antibodies with human and mouse reactivity
ELISABoth unconjugated and conjugated antibodies
Immunohistochemistry (IHC)Rabbit polyclonal antibodies
Immunofluorescence (IF)Rabbit polyclonal antibodies, including FITC-conjugated
Immunoprecipitation (IP)Available for protein interaction studies

These applications enable researchers to detect, localize, and quantify PKD1L2 expression in various experimental contexts .

What are the recommended storage conditions for PKD1L2 antibodies?

For optimal stability and activity, PKD1L2 antibodies should be stored according to the following guidelines:

  • Storage temperature: -20°C for most antibodies; some require -80°C for long-term storage

  • Aliquoting: To avoid repeated freeze-thaw cycles, aliquot antibodies upon receipt

  • Buffer composition: Many PKD1L2 antibodies are supplied in PBS with 0.02% sodium azide and 50% glycerol at pH 7.3

  • Shelf life: Typically 12 months from shipment date when stored properly

  • Shipping conditions: Most suppliers ship these antibodies on blue ice or at 4°C

To minimize performance fluctuations, strict adherence to storage protocols and consistent laboratory conditions are strongly recommended .

How can researchers validate the specificity of PKD1L2 antibodies?

Validating antibody specificity is crucial for reliable experimental results. For PKD1L2 antibodies, the following validation approaches are recommended:

  • Western blot analysis: Verify that the antibody recognizes a band of the expected size (~268 kDa). As demonstrated in research, PKD1L2-specific antibodies (APKD1L2) recognized a ~268 kDa band from protein complexes immunoprecipitated using AFASN antibody in skeletal muscle .

  • Immunoprecipitation followed by mass spectrometry: The immunoprecipitated protein can be analyzed by peptide mass fingerprinting to confirm PKD1L2 identity, as done in studies where "analysis of the corresponding band by peptide mass fingerprinting identified PKD1L2 among other proteins" .

  • Cross-validation with multiple antibodies: Using two different antibodies targeting distinct epitopes of PKD1L2 provides stronger evidence of specificity. For example, researchers validated that "the immunoprecipitated protein was also recognized by the second antibody, APKD1L2_2" .

  • Testing in knockout/knockdown models: Where available, comparing antibody signals between wild-type and PKD1L2-deficient samples provides definitive validation.

  • Recombinant protein controls: Using purified recombinant PKD1L2 protein as a positive control .

What is known about PKD1L2's role in neuromuscular function and disease?

PKD1L2 has been implicated in neuromuscular function and disease through several key findings:

  • Genetic evidence: In an N-ethyl N-nitrosourea-induced mouse mutant called "ostes," upregulation of PKD1L2 was linked to a complex neuromuscular phenotype. This represents "the first role for a TRPP channel in neuromuscular integrity and disease" .

  • Neuromuscular pathology: Ostes mice exhibited chronic neuromuscular impairments including "neuromuscular junction degeneration, polyneuronal innervation and myopathy" .

  • Transgenic confirmation: Ectopic expression of PKD1L2 in transgenic mice reproduced the ostes myopathic changes and caused severe muscle atrophy in Tg(Pkd1l2)/Tg(Pkd1l2) mice, establishing a causal relationship .

  • Dose-dependent effect: Double-heterozygous mice (ostes/+, Tg(Pkd1l2)/0) showed more profound myopathic changes than each heterozygote, indicating a "positive correlation between PKD1L2 levels and disease severity" .

  • Protein interactions: In vivo, PKD1L2 primarily associates with endogenous fatty acid synthase in normal skeletal muscle, and these proteins co-localize to costameric regions of muscle fibers .

  • Metabolic connection: In diseased ostes/ostes muscle, both PKD1L2 and fatty acid synthase are upregulated, and these mice show signs of abnormal lipid metabolism, suggesting a potential mechanistic link .

For researchers investigating neuromuscular disorders, PKD1L2 antibodies are valuable tools for studying expression levels, localization patterns, and protein interactions in both normal and disease states.

How does PKD1L2 functionally relate to other polycystin family proteins?

PKD1L2 shares structural and functional similarities with other polycystin family proteins, particularly PKD1:

  • Protein structure homology: PKD1L2 belongs to the transient receptor potential polycystic (TRPP) channel family, similar to PKD1 . Both contain multiple transmembrane domains and share structural features.

  • Potential regulatory relationships: Research on PKD1 and PKD2 has shown that they interact via C-terminal coiled-coil domains . PKD1 negatively regulates PKD2 expression through protein-protein interaction, as "full-length PC1 that interacts with PC2 via a C-terminal coiled-coil domain regulates PC2... by down-regulating PC2 expression in a dose-dependent manner" . Similar regulatory relationships might exist between PKD1L2 and other family members.

  • Disease relevance: While mutations in PKD1 and PKD2 cause Autosomal Dominant Polycystic Kidney Disease (ADPKD), upregulation of PKD1L2 is associated with neuromuscular disease . This suggests distinct but potentially overlapping pathophysiological roles.

  • Channel function: PKD1L2, like other polycystins, may function as an ion-channel regulator . PKD2 functions as a nonselective cation channel , suggesting potential functional commonalities.

  • Evolutionary relationships: The polycystin family has evolved diverse functions while maintaining core structural features. PKD1L2 represents an important member of this family with specialized functions.

When studying PKD1L2 with antibodies, researchers should consider potential cross-reactivity with other closely related polycystin family members and include appropriate controls.

What experimental approaches are recommended for studying PKD1L2 protein interactions?

Based on previous successful studies, the following approaches are recommended for investigating PKD1L2 protein interactions:

  • Co-immunoprecipitation (Co-IP): This has been effectively used to demonstrate PKD1L2's association with fatty acid synthase. "Western blot analysis using APKD1L2 antibody specifically recognizes a ∼268 kDa band from protein complexes immunoprecipitated using AFASN antibody in skeletal muscle" . The protocol typically involves:

    • Tissue/cell lysis under non-denaturing conditions

    • Immunoprecipitation with anti-PKD1L2 or anti-interacting protein antibody

    • SDS-PAGE separation followed by Western blotting

    • Probing with antibodies against suspected interacting partners

  • Mass spectrometry analysis of immunoprecipitates: After immunoprecipitation with PKD1L2 antibodies, mass spectrometry can identify novel interacting proteins, as demonstrated in studies where "analysis of the corresponding band by peptide mass fingerprinting identified PKD1L2 among other proteins" .

  • Immunofluorescence co-localization: PKD1L2 and fatty acid synthase were shown to "co-localize to costameric regions of the muscle fibre" . This approach involves:

    • Tissue/cell fixation and permeabilization

    • Double immunostaining with PKD1L2 antibody and antibodies against potential interacting proteins

    • Confocal microscopy analysis of co-localization patterns

  • Proximity ligation assay (PLA): This technique can detect protein interactions with high sensitivity and specificity in situ.

  • Pull-down assays with recombinant proteins: Using purified recombinant PKD1L2 domains to identify direct binding partners.

When designing these experiments, researchers should carefully select antibodies with validated specificity and appropriate controls to ensure reliable results.

What are the best approaches for quantifying PKD1L2 expression changes in disease models?

For accurate quantification of PKD1L2 expression changes in disease models, researchers should consider these methodological approaches:

  • Western blot analysis: This has been successfully used to demonstrate PKD1L2 upregulation in disease models. "PKD1L2 is strongly accumulated in ostes/ostes skeletal muscle compared with wild-type" and "accumulation was observed in both soluble and membrane fractions" . For optimal quantification:

    • Include loading controls (housekeeping proteins)

    • Analyze multiple biological replicates

    • Use standard curves with recombinant protein for absolute quantification

    • Perform densitometric analysis with appropriate software

  • ELISA: Quantitative ELISA kits are available for PKD1L2 with detection ranges of 0.156-10 ng/ml . These are particularly useful for:

    • Processing multiple samples simultaneously

    • High-throughput screening

    • Obtaining precise quantitative data

  • Immunohistochemistry with digital image analysis: This allows quantification of PKD1L2 expression while preserving tissue architecture and cellular context.

  • Subcellular fractionation: As demonstrated in research where PKD1L2 expression was assessed in "both soluble and membrane fractions" , this approach enables quantification of PKD1L2 in different cellular compartments.

  • Comparative analysis in transgenic models: Studies showed that "double-heterozygous mice (ostes/+, Tg(Pkd1l2)/0) suffer from myopathic changes more profound than each heterozygote, indicating positive correlation between PKD1L2 levels and disease severity" . This demonstrates the value of using genetic models with varying expression levels.

When interpreting results, researchers should consider that PKD1L2 expression may vary across different tissues and cellular compartments, necessitating comprehensive analysis.

What controls should be included when using PKD1L2 antibodies in experimental protocols?

To ensure reliable and interpretable results when using PKD1L2 antibodies, researchers should include the following controls:

  • Positive controls:

    • Tissues known to express PKD1L2 (skeletal muscle has been well-documented)

    • Cell lines with confirmed PKD1L2 expression (HeLa cells have been used)

    • Recombinant PKD1L2 protein or overexpression systems

  • Negative controls:

    • Primary antibody omission

    • Isotype control antibodies (matching the host species and isotype of the PKD1L2 antibody)

    • When available, tissues or cells with PKD1L2 knockdown/knockout

    • Peptide competition/blocking experiments using the immunizing peptide

  • Specificity controls:

    • Multiple antibodies targeting different epitopes of PKD1L2 (e.g., APKD1L2_1 and APKD1L2_2 as used in published research)

    • Analysis of band size in Western blots (expected ~268 kDa)

    • Cross-reactivity assessment with related proteins (PKD1, PKD1L1)

  • Procedural controls:

    • Loading controls for Western blots (housekeeping proteins)

    • Standardized protein quantification methods

    • Inclusion of molecular weight markers

Including these controls will help validate antibody specificity, optimize experimental conditions, and ensure accurate interpretation of results when investigating PKD1L2 expression and function.

How can researchers optimize immunohistochemistry protocols for PKD1L2 detection in tissue samples?

Optimizing immunohistochemistry (IHC) protocols for PKD1L2 detection requires careful consideration of several factors:

  • Tissue fixation and processing:

    • Fixation method: Paraformaldehyde (4%) is commonly used for preserving PKD1L2 epitopes

    • Section thickness: 5-7 μm sections typically provide optimal results

    • Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) may be necessary to unmask epitopes

  • Antibody selection and optimization:

    • Choose antibodies specifically validated for IHC (e.g., ABIN7163824 has been validated for IHC applications)

    • Optimize antibody concentration through titration experiments (typically starting at 1-5 μg/ml)

    • Consider using polymer detection systems for enhanced sensitivity

  • Blocking and background reduction:

    • Use 5-10% normal serum from the same species as the secondary antibody

    • Include 0.1-0.3% Triton X-100 for improved antibody penetration

    • Consider using commercial background reducing agents if non-specific staining occurs

  • Detection system selection:

    • DAB (3,3'-diaminobenzidine) for brightfield microscopy

    • Fluorescent secondary antibodies for immunofluorescence, particularly useful for co-localization studies

    • Tyramide signal amplification for detecting low-abundance PKD1L2

  • Counterstaining and mounting:

    • Hematoxylin for nuclear counterstaining in brightfield microscopy

    • DAPI for nuclear counterstaining in fluorescence microscopy

    • Use mounting media appropriate for the detection method

  • Optimization strategies:

    • Perform time course experiments for primary antibody incubation (overnight at 4°C often yields optimal results)

    • Test different antibody diluents to improve signal-to-noise ratio

    • Consider using automated IHC platforms for consistency across experiments

When optimizing these protocols, researchers should systematically modify one variable at a time while maintaining appropriate controls to determine the optimal conditions for PKD1L2 detection in their specific tissue samples.

What are the critical factors to consider when selecting a PKD1L2 antibody for specific research applications?

When selecting a PKD1L2 antibody for a specific research application, researchers should evaluate:

  • Target epitope and antibody specificity:

    • N-terminal antibodies (e.g., those targeting AA 1-306) may detect different isoforms than C-terminal antibodies

    • Consider the specific domain of interest (transmembrane, PLAT domain, etc.)

    • Evaluate cross-reactivity with related proteins (PKD1, PKD1L1, etc.)

  • Host species and antibody format:

    • Available options include rabbit polyclonal , mouse monoclonal , and others

    • Consider host species compatibility with other antibodies for co-staining experiments

    • Evaluate clonality (monoclonal for higher specificity, polyclonal for stronger signal)

  • Validated applications:

    • Ensure the antibody has been validated for your specific application:

      ApplicationRecommended Antibody Examples
      Western BlotABIN7269451, SAB1402653, SAB1401957
      ELISAMultiple options available with validation data
      IHCABIN7163824 (validated with 3 validations)
      IFABIN7163824
  • Species reactivity:

    • Match the antibody's reactivity to your experimental system:

      • Human-reactive: Multiple options including ABIN7163824, ABIN530265

      • Mouse-reactive: ABIN7258234, PSC-15-804

      • Cross-reactive antibodies may be useful for comparative studies

  • Validation evidence:

    • Review validation data provided by manufacturers

    • Check for published literature using the specific antibody

    • Consider the extent of validation (number of validation experiments)

  • Technical specifications:

    • Available conjugates (unconjugated, FITC, biotin, HRP)

    • Formulation and buffer compatibility with your protocols

    • Concentration and working dilution recommendations

By systematically evaluating these factors, researchers can select the most appropriate PKD1L2 antibody for their specific experimental needs, resulting in more reliable and reproducible data.

How can PKD1L2 antibodies be used to investigate its potential role in ion channel regulation?

PKD1L2 has been implicated as an ion-channel regulator , and antibodies can be valuable tools for investigating this function:

  • Co-immunoprecipitation studies:

    • PKD1L2 antibodies can be used to isolate protein complexes and identify associated ion channel proteins

    • Western blot analysis of immunoprecipitates can reveal direct interactions with known channel proteins

    • Mass spectrometry analysis of immunoprecipitates can identify novel channel interactions

  • Functional assays combined with antibody treatments:

    • Patch-clamp electrophysiology in cells/tissues with and without PKD1L2 antibody application

    • Calcium imaging experiments in combination with PKD1L2 antibody treatment

    • Channel activity measurements after immunodepletion of PKD1L2

  • Subcellular localization studies:

    • Immunofluorescence to determine co-localization of PKD1L2 with ion channels in plasma membrane, primary cilia, or other compartments

    • Immuno-electron microscopy for high-resolution localization

    • Membrane fractionation followed by Western blotting to quantify PKD1L2 distribution

  • Domain-specific antibodies:

    • Antibodies targeting specific domains of PKD1L2 can help determine which regions are critical for channel interaction

    • Function-blocking antibodies targeting external epitopes can potentially modulate channel activity

  • Expression correlation studies:

    • Quantitative analysis of PKD1L2 expression in relation to ion channel activity in different tissues or disease states

    • Comparison with other polycystin family members known to function as ion channels

By employing these approaches, researchers can gain insights into PKD1L2's role in ion channel regulation and potentially identify novel therapeutic targets for related disorders.

What methodological approaches can be used to study the relationship between PKD1L2 and lipid metabolism?

Research has shown that PKD1L2 associates with fatty acid synthase in skeletal muscle and that abnormal lipid metabolism is observed in PKD1L2-overexpressing mice . To further investigate this relationship, researchers can employ:

  • Protein-protein interaction studies:

    • Co-immunoprecipitation using PKD1L2 antibodies followed by detection of lipid metabolism enzymes

    • Proximity ligation assay (PLA) to visualize interactions between PKD1L2 and fatty acid synthase in situ

    • FRET/BRET analysis with fluorescently tagged proteins to study dynamic interactions

  • Subcellular localization analysis:

    • Immunofluorescence co-localization of PKD1L2 with lipid droplets, endoplasmic reticulum, or mitochondria

    • Subcellular fractionation followed by Western blotting to quantify PKD1L2 distribution in lipid-rich compartments

    • Live-cell imaging of fluorescently tagged PKD1L2 during lipid metabolism perturbations

  • Metabolic profiling:

    • Lipidomics analysis in tissues with normal versus altered PKD1L2 expression

    • Fatty acid oxidation assays in cells with PKD1L2 knockdown/overexpression

    • Incorporation of labeled fatty acids in PKD1L2-manipulated models

  • Functional assays:

    • Measurement of fatty acid synthase activity in the presence/absence of PKD1L2

    • Analysis of lipid storage and mobilization in models with altered PKD1L2 expression

    • Assessment of mitochondrial function in relation to PKD1L2 expression levels

  • Tissue-specific analysis:

    • Comparison of PKD1L2-lipid metabolism connections across different tissues (muscle, liver, kidney)

    • Analysis of PKD1L2 expression in metabolic disease models

    • Correlation of PKD1L2 levels with lipid profiles in clinical samples

These methodological approaches can help elucidate the functional significance of the PKD1L2-fatty acid synthase interaction and potentially identify new therapeutic targets for disorders involving both neuromuscular function and lipid metabolism.

How can researchers effectively use PKD1L2 antibodies in comparative studies across different species?

To effectively use PKD1L2 antibodies in cross-species comparative studies, researchers should consider:

  • Selection of cross-reactive antibodies:

    • Some PKD1L2 antibodies show reactivity to both human and mouse proteins

    • Epitope conservation analysis can predict potential cross-reactivity

    • Preliminary validation in each species is essential before comparative studies

  • Epitope selection strategies:

    • Target highly conserved regions of PKD1L2 for maximum cross-species reactivity

    • The N-terminal region (AA 1-306) is frequently used for generating antibodies with potential cross-reactivity

    • Sequence alignment tools can identify conserved epitopes across species

  • Validation methods for cross-species studies:

    • Western blot analysis in multiple species to confirm band size and specificity

    • Positive control tissues from each species (skeletal muscle is recommended)

    • Peptide competition assays using species-specific peptides

  • Application-specific considerations:

    • For immunohistochemistry: Optimize fixation and antigen retrieval for each species

    • For Western blotting: Adjust lysis conditions for tissue-specific and species-specific differences

    • For immunoprecipitation: Test antibody binding efficiency in each species

  • Quantitative comparative approaches:

    • Use recombinant protein standards from each species for absolute quantification

    • Apply normalization strategies appropriate for cross-species comparisons

    • Consider evolutionary differences in protein expression patterns

  • Documentation and reporting:

    • Clearly document antibody validation data for each species

    • Report species-specific optimizations in methods sections

    • Acknowledge limitations in cross-species reactivity

By implementing these strategies, researchers can effectively use PKD1L2 antibodies to conduct meaningful comparative studies that advance our understanding of PKD1L2 function and evolution across different species.

What are the considerations for using PKD1L2 antibodies in potential therapeutic research?

While direct therapeutic applications of PKD1L2 antibodies are not yet established, researchers exploring their potential in therapeutic research should consider:

  • Target accessibility and specificity:

    • PKD1L2 contains both intracellular and extracellular domains

    • Antibodies targeting extracellular epitopes would be most suitable for in vivo applications

    • Specificity testing against related polycystin family members is crucial

  • Functional modulation potential:

    • Screening for antibodies that can modulate PKD1L2 activity (activating or inhibiting)

    • Evaluation of antibodies that might disrupt protein-protein interactions, such as PKD1L2-fatty acid synthase binding

    • Assessment of impact on downstream signaling pathways

  • In vitro assessment approaches:

    • Cell-based assays to evaluate antibody internalization

    • Functional assays to assess effects on ion channel activity

    • Cytotoxicity and off-target effect evaluation

  • In vivo considerations:

    • Blood-brain barrier penetration (particularly for neuromuscular applications)

    • Antibody humanization for potential clinical translation

    • Tissue distribution studies using labeled antibodies

  • Disease model selection:

    • The "ostes" mouse model with PKD1L2 upregulation provides a relevant platform

    • Transgenic models with ectopic PKD1L2 expression can be valuable for therapeutic testing

    • Development of additional models representing human conditions

  • Biomarker potential:

    • Evaluation of PKD1L2 as a biomarker in neuromuscular disorders

    • Development of quantitative assays using validated antibodies

    • Correlation of PKD1L2 levels with disease progression

  • Technical antibody modifications:

    • Antibody fragments (Fab, scFv) for improved tissue penetration

    • Antibody-drug conjugates for targeted therapy

    • Bispecific antibodies targeting PKD1L2 and disease-relevant partners

These considerations provide a framework for researchers exploring the potential therapeutic applications of PKD1L2 antibodies, particularly in the context of neuromuscular disorders associated with PKD1L2 dysregulation.

What are common issues when using PKD1L2 antibodies in Western blot applications and how can they be resolved?

Researchers may encounter several challenges when using PKD1L2 antibodies for Western blotting:

  • Difficulty detecting the full-size protein (~268 kDa):

    • Problem: PKD1L2 is a large protein (~268 kDa) that can be difficult to transfer efficiently.

    • Solutions:

      • Use low percentage gels (3-8% gradient gels have been successful)

      • Extend transfer time or use specialized transfer systems for large proteins

      • Consider wet transfer methods with lower methanol concentration

      • Use PVDF membranes instead of nitrocellulose for better retention of large proteins

  • Multiple bands or non-specific binding:

    • Problem: Some PKD1L2 antibodies may recognize multiple bands.

    • Solutions:

      • Optimize antibody concentration (titration experiments)

      • Increase blocking time and concentration

      • Use alternative blocking agents (5% milk vs. BSA)

      • Include peptide competition controls to identify specific bands

      • Consider more stringent washing conditions

  • Weak or no signal:

    • Problem: PKD1L2 may be expressed at low levels in some tissues.

    • Solutions:

      • Enrich for PKD1L2 through immunoprecipitation before Western blotting

      • Use enhanced chemiluminescence or fluorescent detection systems

      • Increase protein loading (up to 50-100 μg per lane)

      • Optimize primary antibody incubation (overnight at 4°C often yields better results)

      • Consider membrane fractionation to concentrate PKD1L2

  • Degradation products:

    • Problem: PKD1L2 may be susceptible to proteolytic degradation.

    • Solutions:

      • Include complete protease inhibitor cocktails in lysis buffers

      • Process samples rapidly and maintain cold conditions

      • Consider direct lysis in Laemmli buffer for immediate denaturation

      • Avoid repeated freeze-thaw cycles of samples

  • Inconsistent results across experiments:

    • Problem: Variability in PKD1L2 detection between experiments.

    • Solutions:

      • Standardize lysate preparation methods

      • Use positive controls (tissues known to express PKD1L2, like skeletal muscle)

      • Implement consistent sample handling protocols

      • Consider using loading controls specific for membrane proteins

By systematically addressing these common issues, researchers can optimize Western blot protocols for more reliable and consistent detection of PKD1L2.

What strategies can help overcome challenges in detecting PKD1L2 in immunofluorescence applications?

Immunofluorescence detection of PKD1L2 can present specific challenges that researchers can address with these strategies:

  • Low signal intensity:

    • Problem: PKD1L2 may be expressed at low levels or in specific subcellular compartments.

    • Solutions:

      • Use signal amplification systems (tyramide signal amplification, HRP-conjugated secondaries)

      • Optimize fixation methods to preserve epitopes (4% PFA is often effective)

      • Try different antigen retrieval methods (heat-induced, enzymatic)

      • Increase primary antibody concentration and incubation time

      • Use high-sensitivity detection systems and cameras

  • High background fluorescence:

    • Problem: Non-specific binding leading to background signal.

    • Solutions:

      • Optimize blocking conditions (longer blocking, different blocking agents)

      • Include 0.1-0.3% Triton X-100 for better antibody penetration

      • Use directly conjugated primary antibodies to eliminate secondary antibody background

      • Include autofluorescence quenching steps (Sudan Black B, TrueBlack)

      • Perform careful and extended washing steps

  • Antibody specificity concerns:

    • Problem: Determining if the observed signal is truly PKD1L2.

    • Solutions:

      • Include appropriate controls (peptide competition, PKD1L2 knockdown if available)

      • Use multiple antibodies targeting different epitopes

      • Correlate immunofluorescence with other detection methods (Western blot)

      • Perform co-localization studies with known PKD1L2 interactors (e.g., fatty acid synthase)

  • Subcellular localization challenges:

    • Problem: Distinguishing membrane vs. cytoplasmic localization.

    • Solutions:

      • Use membrane markers for co-localization (Na+/K+-ATPase, WGA)

      • Perform subcellular fractionation followed by Western blotting to complement IF data

      • Consider super-resolution microscopy for detailed localization

      • Use Z-stack imaging to capture the full three-dimensional distribution

  • Tissue-specific optimization:

    • Problem: Different tissues may require different protocols.

    • Solutions:

      • Optimize fixation times for each tissue type

      • Adjust permeabilization conditions based on tissue density

      • Consider tissue-specific autofluorescence reduction methods

      • Test different section thicknesses for optimal antibody penetration

By implementing these strategies, researchers can enhance the detection of PKD1L2 in immunofluorescence applications, enabling more detailed studies of its subcellular localization and co-localization with interaction partners.

How can researchers address PCR amplification difficulties when studying the PKD1L2 gene?

While this question pertains more to gene analysis than antibody applications, it's relevant for researchers comprehensively studying PKD1L2. Based on the search results, specific difficulties have been reported in PCR amplification of PKD1L2:

  • Amplification challenges reported in literature:

    • In the "ostes" mouse model study, researchers noted that "30 sets of primers in a large number of combinations failed to amplify overlapping segments of the large Pkd1l2 cDNA from several tissues including the liver, kidney and muscle; therefore, a complete sequence scan for this particular cDNA was not achieved" .

    • Similarly, they reported that "a full-length Pkd1l2 cDNA could not be amplified" .

  • Potential causes and solutions:

    • Complex gene structure:

      • Design primers for smaller, manageable fragments rather than attempting full-length amplification

      • Use specialized polymerases designed for GC-rich or structurally complex templates

      • Consider long-range PCR techniques with specialized buffer systems

    • Secondary structure complications:

      • Include additives that reduce secondary structure (DMSO, betaine, glycerol)

      • Optimize denaturation temperature and time

      • Use higher annealing temperatures to reduce non-specific binding

    • Low expression levels:

      • Enrich for PKD1L2 transcripts using targeted capture approaches

      • Increase PCR cycle numbers (with appropriate controls for error rates)

      • Use nested PCR approaches for increased sensitivity

  • Alternative approaches:

    • BAC-based strategies: The researchers successfully used a BAC (RP23-269J16) containing "the full-length genomic sequence of Pkd1l2 and the small flanking genes Bcmo1 and Gcsh" for their transgenic work .

    • Next-generation sequencing: The researchers employed "commercial next-generation sequencing approach (NGS) to sequence all exons in the interval" .

    • Targeted RNA-seq: This can provide transcriptome data without relying on PCR amplification of the complete transcript.

    • Protein-level analysis: When gene analysis is challenging, focusing on protein expression using validated antibodies can provide valuable information.

  • Documentation recommendations:

    • Thoroughly document all attempted primer combinations and PCR conditions

    • Report both successful and unsuccessful approaches to guide future researchers

    • Consider publishing detailed protocols when successful amplification is achieved

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.