KLC1 Antibody

Shipped with Ice Packs
In Stock

Description

KLC1 antibodies have been instrumental in elucidating roles of KLC1 in:

  • Transendothelial Migration (TEM): KLC1 isoform variant 1 (KLC1C) directs the lateral border recycling compartment (LBRC) to sites of leukocyte TEM, a process critical in inflammatory responses .

  • Oncogenic Fusion Proteins:

    • KLC1-ROS1 Fusion: Drives glioma progression via constitutive activation of the JAK2-STAT3 pathway, enhancing cell proliferation, invasion, and chemoresistance .

    • KLC1-ALK Fusion: Identified in lung cancer, this fusion exhibits transforming potential in vitro and is detectable in formalin-fixed paraffin-embedded (FFPE) tissues .

Mechanistic Insights

  • Targeted Recycling: Knockdown of KLC1C disrupts LBRC recycling, reducing TEM efficiency by ~60% .

  • Pathway Activation: KLC1-ROS1 fusion increases JAK2 and STAT3 phosphorylation by >400% compared to wild-type ROS1, upregulating oncogenic targets like c-Myc and MMP-2 .

Therapeutic Implications

  • Cancer Biomarker: KLC1 fusions (e.g., ROS1, ALK) are actionable targets; inhibitors of ROS1/JAK2 synergize with temozolomide to suppress glioma growth .

  • Diagnostic Utility: KLC1-ALK detection in FFPE tissues expands access to fusion screening in clinical settings .

Experimental Optimization

  • Dilution Guidelines:

    • WB: 1:500–1:1000

    • IHC/IF: Manufacturer-specific protocols recommended .

  • Sample Compatibility: Validated in human brain tissue (293T, MCF7, U-87MG cells) and rodent models .

Limitations and Future Directions

  • Species Cross-Reactivity: Most antibodies are optimized for human samples; rodent studies may require validation .

  • Fusion-Specific Assays: Current antibodies detect wild-type KLC1 but may not distinguish fusion isoforms without additional validation .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
Kinesin 2 60/70kDa antibody; Kinesin light chain 1 antibody; Kinesin2 antibody; KLC 1 antibody; KLC antibody; Klc1 antibody; KLC1_HUMAN antibody; KNS 2 antibody; KNS 2A antibody; KNS2 antibody; KNS2A antibody; Medulloblastoma antigen MU MB 2.50 antibody; MGC15245 antibody
Target Names
KLC1
Uniprot No.

Target Background

Function
Kinesin is a microtubule-associated protein responsible for generating force, potentially playing a crucial role in organelle transport. The light chain may function in connecting cargo to the heavy chain or in modulating its ATPase activity.
Gene References Into Functions
  1. Structural plasticity of the N-terminal capping helix might represent a structural determinant for TPR domain structural versatility in cargo binding. PMID: 29036226
  2. All binary complexes (KLC1:APP, KLC1:JIP1, and APP:JIP1) contain conformations with favorable binding free energies, suggesting that KLC1 and JIP1 may participate in APP transport in Alzheimer's disease patients. PMID: 27891669
  3. BNIP-2 is a kinesin-1 adapter involved in vesicular transportation within the cytoplasm, and its association with cargos depends on the interaction of the CRAL-TRIO domain with membrane phosphatidylserine. PMID: 25472445
  4. The G allele and GG genotype of KLC1 rs8702 were significantly over-represented among cataract patients compared to healthy controls and were associated with an increased odds ratio for cataract development. PMID: 25883527
  5. Dnm1L interacts with KLC1 through the tetratricopeptide repeat domains. PMID: 25082190
  6. Microtubule-bound kinesin-1 and kinesin-3 motor domains were visualized at multiple steps in their ATPase cycles, including their nucleotide-free states, at approximately 7 Å resolution using cryo-electron microscopy. PMID: 25209998
  7. Studies indicate that FEZ1 (fasciculation and elongation protein zeta 1), SCOCO (short coiled-coil protein), and kinesins (kinesin heavy chain) are involved in biological transport processes. PMID: 24116125
  8. The expression levels of KLC1 variant E in brain and lymphocytes were significantly higher in Alzheimer's disease patients. PMID: 24497505
  9. This study provides evidence that the combined effect of three variants within the KLC1 gene may predispose individuals to age-related cataract. PMID: 23776437
  10. For the binding of cargos shared by KLC1, we propose a different site located within the groove but not involving N343. PMID: 22470497
  11. Data suggest that KLC1 is required for normal neural differentiation, ensuring proper metabolism of AD-associated molecules APP and Tau and for the proliferation of neural precursors (NPs). PMID: 22272245
  12. Phosphorylation of KLC1 at serine 460 modulates binding and trafficking of calsyntenin-1. PMID: 21385839
  13. Results do not convincingly support kinesin light chain 1 (KLC1) as a major susceptibility gene in any of the studied diseases, although there is a small effect of KLC1 in relation to cataract. PMID: 19911314
  14. The major binding site for kinesin light chain in kinesin heavy chain has been mapped to residues 789-813 at the C-terminal end of the heavy chain stalk domain. PMID: 12475239
  15. For a single nucleotide polymorphism (G58836C in intron 13) in the kinesin light-chain 1 (KNS2) gene, the association between Alzheimer's disease and the C allele is found to be significant. PMID: 15364413
  16. The KNS2 gene may play a role during the early stages of Alzheimer's disease pathogenesis. PMID: 17611642
  17. The KLC1 gene may be a novel susceptibility gene for age-related cataract. PMID: 17653041
  18. Microtubule-dependent functions of von Hippel-Lindau tumor suppressor are influenced by kinesin-2. PMID: 17825299
  19. The present finding supports the involvement of the cytoskeleton and KNS2 in the development of vascular white matter damage, thereby opening up novel fields in the research into leukariois. PMID: 17977659
  20. The KLC1 56836CC variant exerts a significant protective effect on the occurrence of multiple sclerosis. PMID: 17999208
  21. The rs8702 variant of the kinesin light chain 1 genotype proved to exert strong amplifying effects on the occurrence and severity of leukoaraiosis in patients with long-lasting poorly controlled severe hypertension. PMID: 19046961
Database Links

HGNC: 6387

OMIM: 600025

KEGG: hsa:3831

STRING: 9606.ENSP00000414982

UniGene: Hs.20107

Protein Families
Kinesin light chain family
Subcellular Location
Cell projection, growth cone. Cytoplasmic vesicle. Cytoplasm, cytoskeleton.
Tissue Specificity
Found in a variety of tissues. Mostly abundant in brain and spine.

Q&A

What is KLC1 and why are antibodies against it important in research?

KLC1 (kinesin light chain 1) is a component of the kinesin motor protein complex that functions as an adapter molecule rather than a motor protein itself. It associates with kinesin heavy chains (KHCs) through its N-terminal domain, while its six tetratricopeptide repeat (TPR) motifs are involved in binding various cargos such as vesicles, mitochondria, and the Golgi complex . KLC1 is primarily localized in cell projections, cytoplasm, and cytoskeletal structures .

Antibodies against KLC1 are important research tools because they enable scientists to:

  • Detect and quantify KLC1 expression in different tissues and cell types

  • Study the subcellular localization of KLC1 using immunofluorescence techniques

  • Isolate KLC1-containing protein complexes through immunoprecipitation

  • Examine changes in KLC1 expression or localization in disease states

  • Investigate post-translational modifications of KLC1, such as phosphorylation

The availability of specific antibodies allows researchers to elucidate the mechanisms of intracellular transport and the role of kinesin proteins in various biological processes, particularly in neuroscience, where axonal transport is critical for neuronal function .

What applications are KLC1 antibodies most commonly used for?

KLC1 antibodies are utilized across multiple experimental applications, with Western blot (WB) being the most widely employed method . Common applications include:

ApplicationFrequency of UseTypical Dilution Range
Western Blot (WB)Very common1:200-1:1000
Immunofluorescence (IF)Common1:50-1:200
Immunohistochemistry (IHC)Common1:50-1:200
ELISACommonVaries by product
Immunoprecipitation (IP)Less commonProduct-dependent
Flow Cytometry (FCM)Less commonProduct-dependent

When selecting a KLC1 antibody, researchers should consider the specific application requirements and choose an antibody that has been validated for their intended use . For instance, certain applications like immunoelectron microscopy may require antibodies with particularly high specificity and affinity characteristics .

How can I validate the specificity of a KLC1 antibody?

Validating antibody specificity is crucial for ensuring reliable experimental results. For KLC1 antibodies, consider the following validation methods:

  • Positive and negative controls: Use tissues or cell lines known to express different levels of KLC1. Mouse brain, rat brain, human brain, and cell lines such as 293T, MCF7, and U-87MG have been validated as positive controls for KLC1 antibodies .

  • Western blot analysis: Look for a single band at approximately 65 kDa, which is the expected molecular weight of KLC1 . Multiple bands might indicate cross-reactivity or detection of different isoforms.

  • Knockdown or knockout validation: Compare antibody signals between wild-type cells and cells where KLC1 has been silenced using siRNA or CRISPR-Cas9 technology .

  • Pre-adsorption tests: Pre-incubate the antibody with purified KLC1 protein before application to your sample. This should eliminate specific staining.

  • Phospho-specific validation: For phospho-specific antibodies (e.g., anti-KLC1 phosphorylated at Thr466), validate specificity by treating samples with phosphatase to remove phosphorylation and confirm signal loss .

  • Phos-tag mobility shift assays: This technique can be used to validate phospho-specific antibodies by separating phosphorylated and non-phosphorylated forms of KLC1 .

Methodologically, a comprehensive validation should include multiple approaches to build confidence in antibody specificity before proceeding with critical experiments.

How do different phosphorylation states of KLC1 affect its interactions with binding partners?

Phosphorylation of KLC1 at specific residues significantly modulates its interactions with cargo proteins and influences kinesin-1-mediated transport. Two key phosphorylation sites that have been characterized include Thr466 and Ser460.

Thr466 Phosphorylation:
The phosphorylation of KLC1 at Thr466 modifies its interaction with JIP1 (JNK-interacting protein 1), a scaffold protein involved in cargo binding. Research has shown that:

  • Unphosphorylated KLC1 preferentially associates with JIP1b through conventional interaction between the TPR domains of KLC1 and the C-terminal region of JIP1b .

  • Phosphorylation at Thr466 suppresses this conventional interaction with JIP1b .

  • This phosphorylation-dependent modulation may serve as a regulatory mechanism for kinesin-1-mediated transport in neurons.

Ser460 Phosphorylation:
Modification of Ser460 affects KLC1's interaction with calsyntenin-1 but not with other cargo proteins:

  • KLC1 with Ser460 mutated to alanine (preventing phosphorylation) binds approximately twofold more calsyntenin-1 than wild-type KLC1 .

  • KLC1 with Ser460 mutated to aspartate (mimicking phosphorylation) binds approximately twofold less calsyntenin-1 than wild-type KLC1 .

  • Importantly, mutation of Ser460 does not alter binding of KLC1 to other cargo proteins such as HAP1A, CRMP2, JIP1, or Kidins220 .

These findings highlight the selective nature of phosphorylation-mediated regulation of KLC1-cargo interactions. Methodologically, researchers investigating these interactions typically employ in vitro pull-down assays, co-immunoprecipitation experiments, and site-directed mutagenesis to create phosphomimetic or phospho-deficient mutants .

What are the optimal experimental conditions for using KLC1 antibodies in phosphorylation studies?

When studying KLC1 phosphorylation, several methodological considerations are critical for obtaining reliable and interpretable results:

Preserving Phosphorylation Status:

  • Include phosphatase inhibitors (e.g., okadaic acid at 400 nM) in lysis buffers to prevent dephosphorylation during sample preparation .

  • For negative controls, treat parallel samples with lambda protein phosphatase (λ PPase) to remove phosphorylation .

  • Process samples quickly and keep them cold throughout to minimize dephosphorylation.

Detection Methods for Phosphorylated KLC1:

  • Phospho-specific antibodies: Use antibodies that specifically recognize KLC1 phosphorylated at sites like Thr466. These should be validated against non-phosphorylated KLC1 to ensure specificity .

  • Phos-tag SDS-PAGE: This technique separates phosphorylated proteins based on their phosphorylation status, allowing visualization of differently phosphorylated forms of KLC1 .

  • Mass spectrometry: LC-MS/MS analysis can identify and quantify phosphorylation sites on KLC1 with high precision. Enrichment steps using IMAC or TiO₂ resin significantly improve detection of phosphopeptides .

Experimental Protocol Example for Phosphorylation Analysis:

  • Treat cells with phosphatase inhibitors (e.g., 400 nM okadaic acid for 3 hours).

  • Lyse cells in buffer containing protease and phosphatase inhibitors.

  • Immunoprecipitate KLC1 using specific antibodies.

  • Analyze by Western blot using phospho-specific antibodies or Phos-tag SDS-PAGE.

  • For comprehensive phosphorylation site mapping, digest immunoprecipitated KLC1 with trypsin and analyze by LC-MS/MS after phosphopeptide enrichment .

This methodological approach ensures accurate detection and characterization of KLC1 phosphorylation events, which is crucial for understanding their functional significance in kinesin-mediated transport.

How can KLC1 antibodies be used to investigate KLC1's role in viral infections?

Recent research has revealed that KLC1 interacts with viral proteins and may serve as a susceptibility factor for certain viral infections, such as dengue virus (DENV). Methodological approaches using KLC1 antibodies to investigate these interactions include:

Co-immunoprecipitation and Proximity Ligation Assays:

  • KLC1 antibodies can be used to immunoprecipitate KLC1 along with associated viral proteins (e.g., DENV NS1) from infected cells .

  • Proximity ligation assays using KLC1 antibodies in conjunction with antibodies against viral proteins can detect protein-protein interactions in situ with high specificity and sensitivity .

Transmission Immunoelectron Microscopy:

  • KLC1 antibodies conjugated to gold particles allow visualization of KLC1 localization relative to viral structures at ultrastructural resolution.

  • This technique has revealed KLC1 decorating the surface of vacuoles in association with viral proteins like NS1 .

Silencing Studies:

  • After silencing KLC1 expression (e.g., using siRNA), KLC1 antibodies can be used to confirm knockdown efficiency by Western blot or immunofluorescence.

  • Subsequent analysis of viral replication provides insights into KLC1's functional role in the viral life cycle .

Temporal Expression Analysis:

  • KLC1 antibodies enable tracking of changes in KLC1 expression levels during the course of viral infection.

  • For example, increased levels of KLC1 were observed starting at 6 hours post-infection with DENV, suggesting that the virus stimulates KLC1 synthesis .

A comprehensive experimental approach would combine these methods to determine both the physical interactions between KLC1 and viral components and the functional significance of these interactions for viral replication. This research direction has significant implications for understanding viral pathogenesis and potentially developing novel antiviral strategies.

What are the considerations for selecting KLC1 antibodies for cross-species studies?

When conducting cross-species studies involving KLC1, careful selection of antibodies is essential to ensure reliable detection across different species. Consider the following methodological approaches:

Epitope Conservation Analysis:

  • Align KLC1 protein sequences from target species to identify conserved regions.

  • Select antibodies raised against epitopes within highly conserved regions for maximum cross-reactivity.

  • Avoid antibodies targeting regions with species-specific sequence variations.

Validated Cross-Reactivity:
Many commercial KLC1 antibodies have been validated for cross-reactivity with multiple species:

Antibody TypeValidated Species ReactivityApplicationsReference
Rabbit MonoclonalHuman, Mouse, RatWB, ELISA
Rabbit PolyclonalHuman, Mouse, RatWB, IHC
Mouse Monoclonal (L2)Human, Mouse, RatWB, IP, IF, ELISA

Additionally, some antibodies have been validated for more specialized model organisms:

  • Drosophila-specific KLC antibodies for use in insect models

  • C. elegans-specific klc-2 antibodies for nematode studies

Experimental Validation Strategy:

  • Positive controls: Include samples from species with known antibody reactivity.

  • Western blot analysis: Confirm the antibody detects bands of appropriate molecular weight across all species of interest.

  • Preabsorption controls: Perform with recombinant KLC1 proteins from different species.

  • Dilution optimization: Optimal antibody dilutions may vary between species; perform dilution series for each species.

This methodological approach enables robust cross-species studies of KLC1, which is particularly valuable for evolutionary studies, comparative physiology, and validation of disease models across different species .

How can phospho-specific KLC1 antibodies be developed and validated?

Development and validation of phospho-specific antibodies against KLC1 requires a systematic approach to ensure specificity and sensitivity:

Development Process:

  • Phosphopeptide design: Create synthetic peptides containing the phosphorylated residue of interest (e.g., C+TVTTpTLKNL-CONH₂ for KLC1 phosphorylated at Thr466) .

  • Immunization: Immunize host animals (typically rabbits) with the phosphopeptide conjugated to a carrier protein.

  • Antibody purification: Perform serial affinity chromatography:

    • First, purify antibodies using columns with the phosphorylated peptide

    • Then, remove antibodies that recognize non-phosphorylated epitopes by passing through columns containing the non-phosphorylated peptide

    • Finally, remove antibodies that recognize phosphothreonine/phosphoserine alone

Rigorous Validation Protocol:

  • Western blot comparison: Compare antibody reactivity against phosphorylated vs. dephosphorylated (phosphatase-treated) samples .

  • Phos-tag mobility shift assays: Verify that the antibody recognizes the shifted (phosphorylated) band in Phos-tag SDS-PAGE .

  • Phosphomimetic mutants: Test reactivity against wild-type protein vs. phosphomimetic (e.g., Ser/Thr to Asp/Glu) and phospho-deficient (e.g., Ser/Thr to Ala) mutants.

  • Mass spectrometry confirmation: Confirm phosphorylation at the expected site in immunoprecipitated samples using LC-MS/MS .

  • Immunocytochemistry with controls: Perform immunostaining of cells with known phosphorylation status (e.g., okadaic acid-treated vs. phosphatase-treated).

Application in Research:
Properly validated phospho-specific KLC1 antibodies enable:

  • Temporal monitoring of phosphorylation events during cellular processes

  • Spatial mapping of phosphorylated KLC1 within cells

  • Quantification of phosphorylation levels under different experimental conditions

  • Identification of stimuli that trigger KLC1 phosphorylation

This methodological approach has successfully produced antibodies specific to KLC1 phosphorylated at Thr466, contributing to our understanding of how phosphorylation regulates kinesin-cargo interactions .

What are the optimal fixation and permeabilization conditions for KLC1 immunocytochemistry?

For successful immunocytochemical detection of KLC1, appropriate fixation and permeabilization protocols are essential to preserve protein structure while allowing antibody access. Based on published protocols:

Fixation Recommendations:

  • Paraformaldehyde fixation: 4% paraformaldehyde in PBS for 15-20 minutes at room temperature preserves KLC1 structure while maintaining cellular architecture.

  • Methanol fixation: Ice-cold methanol for 10 minutes at -20°C can provide good results for certain applications, particularly when examining cytoskeletal associations.

  • Avoid formaldehyde-glutaraldehyde mixtures for standard immunofluorescence as these can mask KLC1 epitopes.

Permeabilization Options:

  • Triton X-100: 0.1-0.5% in PBS for 5-10 minutes is commonly used and works well for accessing cytoplasmic KLC1.

  • Saponin: 0.1% provides gentler permeabilization that better preserves membrane structures when examining KLC1 associated with vesicles.

  • Digitonin: 40 µg/ml can be used for selective plasma membrane permeabilization when studying KLC1 in relation to organelle membranes.

Protocol Optimization Tips:

  • Test multiple conditions as the optimal protocol may vary depending on cell type and the specific KLC1 antibody used.

  • For phospho-specific KLC1 antibodies, include phosphatase inhibitors (e.g., 10 mM sodium fluoride, 1 mM sodium orthovanadate) in all buffers.

  • Blocking with 5% normal serum from the species of the secondary antibody for 30-60 minutes reduces background staining.

  • For double labeling with microtubule markers, methanol fixation may provide superior results as it preserves microtubule structures.

These methodological considerations help optimize KLC1 detection while preserving its native localization patterns within cells, which is critical for studies of intracellular transport processes.

How should KLC1 antibodies be stored and handled to maintain optimal activity?

Proper storage and handling of KLC1 antibodies is crucial for maintaining their specificity and sensitivity over time. Based on manufacturer recommendations and research practices:

Storage Conditions:

  • Temperature: Store most KLC1 antibodies at -20°C for long-term storage. Avoid repeated freeze-thaw cycles by making small aliquots .

  • Buffer composition: Most commercial KLC1 antibodies are supplied in buffers containing:

    • Phosphate buffered solution (pH 7.4)

    • Stabilizers (typically 0.05%)

    • Glycerol (often 50%) to prevent freezing and denaturation

  • Conjugated antibodies: Fluorophore-conjugated KLC1 antibodies (FITC, PE, Alexa Fluor®) should be protected from light during storage to prevent photobleaching .

Handling Practices:

  • Working dilutions: Prepare fresh working dilutions for each experiment rather than storing diluted antibody.

  • Thawing: Thaw frozen antibodies completely before use, then mix gently to ensure homogeneity. Avoid vortexing, which can denature antibodies.

  • Temperature transitions: Allow refrigerated antibodies to equilibrate to room temperature before opening to prevent condensation, which can introduce contaminants.

  • Centrifugation: Briefly centrifuge antibody vials before use to collect all liquid at the bottom of the tube.

Stability Considerations:

  • Most KLC1 antibodies remain stable for approximately 12 months when stored properly .

  • Some antibody formulations (particularly agarose-conjugated ones) may have shorter shelf lives or specific storage requirements .

  • If decreased activity is observed over time, validation experiments (e.g., Western blot with positive control samples) should be performed to assess antibody performance.

Following these storage and handling recommendations helps ensure consistent and reliable results in experiments using KLC1 antibodies, particularly for quantitative applications where antibody performance is critical.

What controls should be included when using KLC1 antibodies in co-immunoprecipitation experiments?

Co-immunoprecipitation (Co-IP) experiments using KLC1 antibodies require rigorous controls to ensure valid and interpretable results, particularly when investigating novel KLC1 binding partners:

Essential Controls for KLC1 Co-IP:

  • Input control:

    • Include 5-10% of the lysate used for IP to verify the presence of both KLC1 and potential interacting proteins before immunoprecipitation.

    • This control establishes baseline expression levels and confirms that proteins are detectable by the antibodies used.

  • Negative/isotype control:

    • Perform parallel IP with an isotype-matched irrelevant antibody (same species and isotype as the KLC1 antibody) to identify non-specific binding.

    • For example, if using mouse monoclonal anti-KLC1 (IgG2a), use a mouse IgG2a antibody against an irrelevant target as control .

  • Bead-only control:

    • Include a sample with beads but no primary antibody to identify proteins that bind non-specifically to the beads.

    • This is particularly important when using Protein A/G beads directly rather than antibody-conjugated beads.

  • Reciprocal IP:

    • If studying a specific KLC1 interaction, perform IP with antibodies against both KLC1 and the proposed interacting protein.

    • Confirming the interaction in both directions significantly strengthens the evidence for a genuine interaction.

  • Phosphatase treatment control:

    • When studying phosphorylation-dependent interactions, include samples treated with lambda phosphatase to demonstrate phosphorylation-dependence .

    • This control is critical when using phospho-specific KLC1 antibodies or investigating phosphorylation-dependent interactions.

Experimental Design Considerations:

Control TypePurposeImplementation
InputVerify protein expression5-10% of pre-IP lysate
Isotype controlDetect non-specific bindingNon-relevant antibody of same isotype
Bead-onlyDetect bead-protein interactionsNo primary antibody
Reciprocal IPConfirm bidirectional interactionIP with partner protein antibody
Phosphatase treatmentVerify phospho-dependenceλ-PPase treatment of lysates

These controls help distinguish true interactions from artifacts and provide compelling evidence for physiologically relevant KLC1 binding partners, which is essential for advancing our understanding of kinesin-mediated transport mechanisms .

What are common pitfalls when using KLC1 antibodies in Western blotting and how can they be resolved?

Western blotting with KLC1 antibodies can present several challenges. Here are common issues and their methodological solutions:

Issue: Multiple bands or unexpected molecular weight

Potential causes and solutions:

  • Detection of KLC1 isoforms: KLC1 has multiple splice variants. Review literature to identify expected isoform patterns in your experimental system .

  • Cross-reactivity with other KLC family members: Use antibodies raised against unique KLC1 regions rather than conserved domains.

  • Degradation products: Add fresh protease inhibitors to lysis buffer and keep samples cold throughout preparation.

  • Post-translational modifications: Phosphorylation can cause mobility shifts. Use Phos-tag gels to separate phosphorylated forms or treat samples with phosphatase to confirm .

Issue: Weak or no signal

Potential causes and solutions:

  • Inadequate transfer: Optimize transfer conditions for high molecular weight proteins (~65 kDa).

  • Low expression levels: Increase the amount of loaded protein or use enrichment methods like immunoprecipitation.

  • Epitope masking: Try different sample preparation methods; boiling can sometimes mask epitopes in membrane proteins.

  • Incorrect antibody dilution: Titrate antibody concentration using a positive control sample (e.g., mouse brain, 293T cells) .

  • Improper blocking: Certain blocking agents may interfere with antibody binding; test different blockers (BSA vs. milk).

Issue: High background

Potential causes and solutions:

  • Non-specific binding: Increase washing frequency/duration and optimize antibody dilution.

  • Insufficient blocking: Extend blocking time or increase blocker concentration.

  • Secondary antibody cross-reactivity: Use secondary antibodies specifically adsorbed against cross-reactive species.

  • Membrane contamination: Handle membranes with clean gloves and forceps.

Optimization Strategy:

ParameterStarting ConditionOptimization Range
Antibody Dilution1:5001:200-1:1000
Blocking Agent5% milk3-5% milk or BSA
Incubation TimeOvernight at 4°C1h at RT to overnight at 4°C
Wash BufferTBST (0.1% Tween)Increase Tween to 0.2% for higher stringency
Sample PreparationBoiling in SDS bufferTry non-boiled or partially denatured samples

This methodological approach to troubleshooting ensures reliable and reproducible detection of KLC1 in Western blotting applications, which is critical for quantitative analyses of KLC1 expression or modification .

How can I optimize detection of KLC1 in immunohistochemistry of fixed tissue samples?

Optimizing KLC1 detection in fixed tissue samples requires careful attention to multiple parameters, as KLC1's cytoskeletal and vesicular associations make it susceptible to fixation artifacts. Follow these methodological approaches for successful immunohistochemical staining:

Tissue Fixation Optimization:

  • Fixation method comparison: Test both perfusion and immersion fixation to determine optimal preservation of KLC1 epitopes.

  • Fixative composition: Compare 4% paraformaldehyde in PBS versus paraformaldehyde-lysine-periodate (PLP) fixative, which can better preserve cytoskeletal proteins.

  • Fixation duration: Overfixation can mask epitopes; test shorter fixation times (4-12 hours) for improved antibody access.

Antigen Retrieval Methods:
KLC1 detection often benefits from antigen retrieval to unmask epitopes:

  • Heat-induced epitope retrieval (HIER): Test both citrate buffer (pH 6.0) and Tris-EDTA buffer (pH 9.0) at 95-100°C for 15-20 minutes.

  • Enzymatic retrieval: For some tissues, light protease treatment (0.01% trypsin for 5-10 minutes) may improve antibody access to KLC1.

  • Combined approaches: Sequential application of enzymatic and heat-based methods may yield superior results for difficult samples.

Tissue-Specific Considerations:

  • Nervous tissue: KLC1 is highly expressed in neurons, but detection can be challenging due to lipid content. Extending permeabilization times may improve results.

  • Brain tissues: Include sudan black treatment (0.1% in 70% ethanol for 20 minutes) to reduce lipofuscin autofluorescence when performing immunofluorescence.

  • Highly fixated tissues (e.g., archival samples): More aggressive antigen retrieval and higher antibody concentrations (1:50 dilution) may be necessary .

Protocol Optimization Strategy:

ParameterVariable to TestEvaluation Method
Antibody dilutionRange from 1:50 to 1:200 Signal-to-noise ratio
Incubation time1 hour at RT vs. overnight at 4°CStaining intensity and specificity
Blocking agentNormal serum vs. BSA vs. commercial blockersBackground reduction
Detection systemHRP/DAB vs. fluorescent secondary antibodiesSensitivity and resolution
AmplificationStandard vs. tyramide signal amplificationSignal enhancement for low-abundance targets

This methodological approach has been successfully applied to detect KLC1 in human brain and cervical cancer tissues, demonstrating its practical utility for clinical and research specimens .

How can KLC1 antibodies be used to investigate changes in KLC1 expression during disease progression?

KLC1 has been implicated in several pathological conditions, particularly neurodegenerative diseases and cancer. Using KLC1 antibodies to track expression changes during disease progression requires methodologically sound approaches:

Quantitative Western Blot Analysis:

  • Standardized protein extraction: Use consistent extraction protocols across all disease stages to ensure comparable results.

  • Internal loading controls: Include both general loading controls (β-actin, GAPDH) and subcellular compartment-specific controls (e.g., α-tubulin for cytoskeletal fraction).

  • Quantification method: Use densitometry with linear range validation to ensure measurements fall within the quantifiable range.

  • Statistical analysis: Analyze multiple samples from each disease stage (minimum n=5) to account for individual variation.

Immunohistochemical Analysis of Tissue Progression:

  • Tissue microarrays: Use arrays containing samples from different disease stages to allow for direct comparison under identical staining conditions.

  • Automated image analysis: Employ digital pathology tools to quantify KLC1 staining intensity and distribution.

  • Cell type-specific analysis: Combine KLC1 antibodies with cell type markers to determine if expression changes are global or cell type-specific.

  • Spatial analysis: Evaluate changes in subcellular localization (e.g., cytoplasmic vs. membrane-associated) across disease stages.

Analysis in Model Systems:

  • Temporal sampling: In disease models, collect samples at defined time points to create a temporal profile of KLC1 expression changes.

  • Correlation with disease markers: Correlate KLC1 expression with established biomarkers of disease progression.

  • Intervention studies: Use KLC1 antibodies to assess whether therapeutic interventions normalize KLC1 expression patterns.

Protocol Example for Neurodegenerative Disease Models:

  • Collect brain tissue samples from transgenic models at pre-symptomatic, early symptomatic, and late-stage time points.

  • Process parallel samples for Western blot and immunohistochemistry.

  • Probe Western blots with anti-KLC1 antibodies (1:500 dilution) and quantify relative to loading controls.

  • Perform double immunostaining with anti-KLC1 (1:100) and cell type-specific markers.

  • Analyze changes in both total expression level and subcellular distribution patterns.

  • Correlate findings with behavioral deficits and other pathological markers.

This comprehensive methodological approach enables researchers to establish whether changes in KLC1 expression contribute to pathogenesis or represent compensatory responses in disease progression .

How does the interaction between KLC1 and viral proteins contribute to viral pathogenesis?

Recent research has revealed a previously unknown role for KLC1 in viral infections, particularly with dengue virus (DENV). The methodological approaches used to elucidate these interactions provide valuable insights for researchers studying host-pathogen interactions:

KLC1-NS1 Interaction in Dengue Virus Infection:
A groundbreaking study published in March 2025 demonstrated that KLC1 interacts with the DENV non-structural protein 1 (NS1) and functions as a susceptibility factor for DENV infection in mosquito cells . Key findings include:

  • Temporal dynamics: KLC1 expression increases starting at 6 hours post-infection, suggesting virus-induced upregulation .

  • Physical interaction: The KLC1-NS1 interaction was confirmed using multiple complementary techniques:

    • Proximity ligation assays showing interaction in intact cells

    • Co-immunoprecipitation confirming biochemical association

    • Transmission immunoelectron microscopy revealing KLC1 decorating vacuoles associated with NS1

  • Functional significance: Silencing KLC1 expression disrupted the DENV replication cycle in mosquito cells, establishing KLC1 as a necessary host factor .

Methodological Implications for Viral Research:
These findings suggest a general approach for investigating kinesin involvement in viral infections:

  • Proteomics screening: Identify potential interactions between viral proteins and host transport machinery components.

  • Validation cascade: Confirm interactions through complementary methods (co-IP, proximity ligation, microscopy).

  • Functional testing: Use gene silencing or inhibition to establish the necessity of the interaction for viral replication.

  • Mechanistic investigation: Determine whether the interaction facilitates viral transport, replication complex assembly, or other aspects of the viral life cycle.

The discovery that KLC1 serves as a susceptibility factor for DENV opens new avenues for antiviral development, potentially through disrupting KLC1-viral protein interactions. Similar approaches could be applied to investigate the role of KLC1 in other viral infections, particularly those known to manipulate host cell membranes and trafficking pathways .

What are the cutting-edge techniques being used to study KLC1 post-translational modifications?

Research into KLC1 post-translational modifications (PTMs) has been advanced by several cutting-edge techniques that provide unprecedented insights into the regulation of kinesin-mediated transport:

Mass Spectrometry-Based Approaches:

  • Phosphoproteomics with sequential enrichment: This approach combines IMAC and TiO₂ resin enrichment steps to comprehensively identify phosphorylation sites on KLC1 .

  • Quantitative MS with stable isotope labeling: SILAC or TMT labeling enables quantitative comparison of phosphorylation levels under different conditions.

  • Top-down proteomics: Analysis of intact KLC1 protein to identify combinatorial patterns of multiple PTMs that might be lost in traditional bottom-up approaches.

  • Targeted phosphopeptide MS: Selective reaction monitoring (SRM) or parallel reaction monitoring (PRM) for precise quantification of specific phosphorylated KLC1 peptides.

Advanced Imaging Methods:

  • Super-resolution microscopy: Techniques like STORM and PALM provide nanoscale resolution of KLC1 localization and its relationship to phosphorylation state.

  • FRET-based biosensors: Engineered sensors that change conformation upon KLC1 phosphorylation, enabling real-time monitoring in living cells.

  • Phospho-specific antibody-based proximity ligation: Allows visualization of specific phosphorylated forms of KLC1 in situ with high sensitivity.

Biochemical and Functional Assays:

  • Phos-tag mobility shift assays: Separate phosphorylated from non-phosphorylated KLC1 to quantify phosphorylation stoichiometry .

  • In vitro reconstitution with defined components: Assess how specific phosphorylation events affect cargo binding and motor activity using purified components.

  • Optogenetic control of kinase activity: Allow temporal precision in inducing KLC1 phosphorylation to study immediate functional consequences.

Computational Approaches:

  • Molecular dynamics simulations: Model the structural consequences of phosphorylation on KLC1-cargo interactions.

  • Machine learning algorithms: Predict functional consequences of novel phosphorylation sites based on known examples.

  • Systems biology modeling: Integrate KLC1 phosphorylation into broader signaling networks controlling cellular transport.

These methodological advances are rapidly expanding our understanding of how PTMs, particularly phosphorylation at sites like Thr466 and Ser460, regulate KLC1 function in normal physiology and disease states .

What is the current understanding of KLC1's role in neurodegenerative diseases?

KLC1 plays a crucial role in axonal transport, and disruptions in this process are implicated in several neurodegenerative diseases. Current research using KLC1 antibodies has provided important insights into these connections:

Alzheimer's Disease (AD):

  • Tau-kinesin interactions: Phosphorylated tau protein can interfere with kinesin-based transport. KLC1 antibodies have been used to demonstrate altered KLC1-tau interactions in AD brain samples .

  • APP transport: KLC1 is involved in the transport of amyloid precursor protein (APP). Perturbations in this transport may contribute to amyloidogenic processing.

  • JIP1 scaffolding: The interaction between KLC1 and JIP1 is modulated by phosphorylation at Thr466. This regulatory mechanism appears disrupted in AD models .

Parkinson's Disease (PD):

  • α-synuclein accumulation: Defects in KLC1-mediated transport may contribute to α-synuclein aggregation in dopaminergic neurons.

  • Mitochondrial transport: KLC1 facilitates mitochondrial movement, and impaired mitochondrial transport is a feature of PD pathogenesis.

Methodological Approaches to Study KLC1 in Neurodegeneration:

  • Human tissue analysis: Comparative studies of KLC1 expression, localization, and phosphorylation state between control and diseased brain tissue using KLC1 antibodies.

  • Functional transport assays: Live imaging of cargo movement in neurons expressing wild-type or mutant forms of KLC1.

  • Axonal blockage assessment: Use of KLC1 antibodies to identify axonal swellings containing accumulated KLC1 and cargo proteins.

  • Therapeutic targeting studies: Testing whether normalization of KLC1 function (e.g., through phosphorylation modulators) can restore axonal transport and mitigate disease phenotypes.

Emerging Therapeutic Implications:
Understanding KLC1's role in these diseases suggests several potential therapeutic strategies:

  • Stabilizing kinesin-cargo interactions: Compounds that prevent disruption of KLC1-cargo binding by pathological proteins.

  • Modulating KLC1 phosphorylation: Targeting specific kinases or phosphatases that regulate KLC1 activity.

  • Enhancing kinesin motor function: Small molecules that boost kinesin processivity to overcome transport deficits.

The continued development and application of specific KLC1 antibodies, particularly phospho-specific variants, will be crucial for advancing our understanding of KLC1's role in neurodegenerative processes and for developing targeted therapeutic approaches .

What emerging technologies might enhance the utility of KLC1 antibodies in research?

Several emerging technologies promise to expand the capabilities and applications of KLC1 antibodies in both basic research and clinical settings:

Next-Generation Antibody Engineering:

  • Single-domain antibodies (nanobodies): These smaller antibody fragments derived from camelid antibodies offer superior tissue penetration and can access epitopes inaccessible to conventional antibodies, potentially revealing new aspects of KLC1 biology.

  • Intrabodies: Engineered antibodies that function within living cells could allow real-time tracking of KLC1 activity and interactions.

  • Bispecific antibodies: Simultaneously targeting KLC1 and cargo proteins could provide unique insights into transport complex formation and dynamics.

Advanced Imaging Technologies:

  • Expansion microscopy: Physical expansion of specimens could allow visualization of KLC1-cargo interactions at super-resolution using standard confocal microscopy.

  • Lattice light-sheet microscopy: Enables long-term, high-resolution imaging of KLC1-mediated transport in living cells with minimal phototoxicity.

  • Cryo-electron tomography: Could reveal the structural organization of KLC1 within native transport complexes at near-atomic resolution.

Spatially-Resolved Proteomics:

  • Imaging mass cytometry: Allows simultaneous detection of multiple proteins, including different phosphorylated forms of KLC1, with subcellular resolution in tissue sections.

  • Proximity labeling proteomics: Techniques like BioID or APEX2 fused to KLC1 could map the dynamic interactome of KLC1 in different cellular compartments.

  • Spatial transcriptomics integration: Correlating KLC1 protein distribution with local transcriptional profiles could reveal regulatory relationships.

High-Throughput and AI-Assisted Analysis:

  • Automated phenotypic screening: Using KLC1 antibodies in high-content screening to identify compounds that normalize KLC1 function in disease models.

  • Deep learning image analysis: AI algorithms could extract subtle patterns in KLC1 distribution and phosphorylation state that correlate with disease progression.

  • Digital pathology integration: Automated quantification of KLC1 immunostaining in patient samples could identify novel biomarkers.

These technological advances will likely transform how researchers use KLC1 antibodies, moving beyond simple detection to dynamic, multiparametric analysis of KLC1 function in complex biological systems, with significant implications for understanding both normal physiology and disease mechanisms .

How might KLC1 antibodies contribute to the development of targeted therapeutics?

KLC1 antibodies have significant potential to contribute to the development of targeted therapeutics for diseases involving disrupted intracellular transport, particularly neurodegenerative disorders and certain viral infections:

Drug Target Identification and Validation:

  • Phosphorylation site mapping: KLC1 phospho-specific antibodies can identify disease-specific phosphorylation patterns, revealing potential targets for kinase or phosphatase inhibitors .

  • Cargo interaction profiling: Antibodies that disrupt specific KLC1-cargo interactions can help validate these interfaces as therapeutic targets.

  • Disease-relevant conformations: Conformation-specific antibodies might distinguish between functional and pathological states of KLC1.

Therapeutic Monitoring:

  • Pharmacodynamic biomarkers: Changes in KLC1 phosphorylation or localization detected by antibodies could serve as indicators of drug efficacy.

  • Patient stratification: Patterns of KLC1 expression or modification might identify patient subgroups most likely to respond to specific treatments.

  • Resistance mechanisms: Monitoring changes in KLC1 and its interactions during treatment could reveal adaptation mechanisms.

Antibody-Based Therapeutics:

  • Intrabodies as protein-protein interaction modulators: Engineered antibodies that selectively disrupt pathological KLC1 interactions while preserving normal function.

  • Viral infection intervention: Based on the discovery of KLC1 as a susceptibility factor for dengue virus, antibody-based approaches to disrupt KLC1-NS1 interaction could have antiviral potential .

  • Targeted cargo delivery: KLC1 antibody fragments conjugated to therapeutic cargoes could leverage natural kinesin transport pathways for precise intracellular delivery.

Methodological Framework for KLC1-Targeted Drug Development:

  • Disease-specific characterization: Use KLC1 antibodies to characterize alterations in different diseases (phosphorylation, localization, interactions).

  • Functional consequence analysis: Determine which alterations directly contribute to pathology versus compensatory responses.

  • Target prioritization: Identify the most druggable aspects of altered KLC1 function.

  • Screening platform development: Establish antibody-based assays to screen for compounds that normalize KLC1 function.

  • Mechanism validation: Confirm that compounds act through the intended mechanism using KLC1 antibodies as analytical tools.

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.