CLN3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the order method and location. Please consult your local distributor for specific delivery timelines.
Synonyms
Batten disease protein antibody; Battenin antibody; BTS antibody; Ceroid lipofuscinosis neuronal 3 antibody; Ceroid lipofuscinosis neuronal 3 juvenile (Batten Spielmeyer Vogt disease) antibody; Ceroid lipofuscinosis neuronal 3 juvenile antibody; CLN 3 antibody; CLN3 antibody; CLN3_HUMAN antibody; JNCL antibody; MGC102840 antibody; Protein CLN3 antibody
Target Names
CLN3
Uniprot No.

Target Background

Function
CLN3 antibody plays a crucial role in mediating microtubule-dependent, anterograde transport, connecting the Golgi network, endosomes, autophagosomes, lysosomes, and the plasma membrane. This protein participates in various cellular processes, including:
  • Regulation of lysosomal pH and protein degradation
  • Receptor-mediated endocytosis
  • Autophagy
  • Transport of proteins and lipids from the trans-Golgi network (TGN)
  • Apoptosis
  • Synaptic transmission
CLN3 facilitates protein transport from the TGN to other membrane compartments, including the transport of microdomain-associated proteins to the plasma membrane. It also mediates the transport of IGF2R to the lysosome, where it regulates the release of CTSD, ultimately controlling CTSD maturation and the intracellular processing of APP. Additionally, CLN3 regulates CTSD activity in response to osmotic stress. CLN3 binds galactosylceramide and transports it from the trans-Golgi to the rafts, potentially impacting cell survival by modulating ceramide synthesis. At the plasma membrane, CLN3 regulates actin-dependent processes, including filopodia formation, cell migration, and pinocytosis through the ARF1-CDC42 pathway. It also contributes to cytoskeleton organization by interacting with MYH10 and fodrin, influencing the association of the Na+, K+ ATPase complex with the plasma membrane. CLN3 regulates synaptic transmission in the amygdala, hippocampus, and cerebellum by regulating synaptic vesicle density and their proximity to active zones, modulating short-term plasticity and influencing age-dependent anxious behavior, learning, and memory. CLN3 regulates autophagic vacuole (AV) maturation by modulating trafficking between endocytic and autophagolysosomal/lysosomal compartments, involving vesicle fusion and controlling the degradation process. It also participates in the cellular homeostasis of compounds like water, ions, amino acids, proteins, and lipids in various tissues, particularly in the brain and kidney, by regulating their transport and synthesis.
Gene References Into Functions
  1. This research suggests that Cln3 plays a role in both conventional and unconventional protein secretion. The absence of Cln3 leads to dysregulated secretion during early development, providing the first evidence linking CLN3 function to protein secretion in any system. PMID: 28365442
  2. AAV2-CLN3 effectively restored full-length CLN3 transcript and protein in patient-specific fibroblasts and induced pluripotent stem cell (iPSC)-derived retinal neurons. Subretinal injections of purified AAV2-CLN3 in wild-type mice did not demonstrate any evidence of retinal toxicity. PMID: 27400765
  3. The lysosomal enzyme cathepsin D (CTSD) mediates the proteolytic cleavage of PSAP precursor into saposins A-D. Myc-CLN3 colocalized with CTSD, and CTSD activity decreased with increased myc-CLN3 expression, particularly under hyperosmotic conditions. PMID: 28390177
  4. This study provides the first detailed morphological evaluation of CLN3 patients in the early years following the onset of ocular symptoms. CLN3 is characterized by early degeneration primarily affecting the first and second neurons compared to other macular and generalized retinal dystrophies. PMID: 27486012
  5. The age at onset and natural progression of retinal disease differ significantly between syndromic and nonsyndromic CLN3 disease, potentially linked to genotypic differences. PMID: 28542676
  6. CLN3 knockdown inhibits cell proliferation and induces G0/G1 cell cycle arrest in the A2780 cell line and its drug-resistant sub-lines. PMID: 26299671
  7. This study elucidates the membrane topology of the human CLN3 protein. PMID: 25051496
  8. Heterozygous carriers of CLN3 disease exhibited normal ocular features and vision compared to a control group, contradicting previous suggestions of retinal dysfunction in these individuals. PMID: 25338278
  9. This new model system, allowing for the study of Cln3 function in both single cells and a multicellular organism, coupled with the observation that human CLN3 expression restores abnormalities in Dictyostelium cln3- cells, provides valuable insights. PMID: 25330233
  10. These findings further support the crucial role of the CLN3 protein in intracellular Ca(2+) handling and autophagic pathway flux, establishing a powerful platform for therapeutic screening. PMID: 25878248
  11. CLN3 mutation is associated with neuronal ceroid lipofuscinosis. PMID: 24271013
  12. Genetic testing for CLN3 should be considered in autophagic vacuolar myopathy (AVM) cases with autophagic vacuoles and sarcolemmal features. PMID: 24827497
  13. This study identified CLN3 as a novel disease gene for non-syndromic retinal diseases, supported by five unrelated patient families. PMID: 24154662
  14. CLN3 is involved in the response and adaptation to cellular stress. PMID: 23840424
  15. Protein interaction mapping analysis suggests CLN3 is involved in transmembrane transport, lipid homeostasis, neuronal excitability, and links it to G-protein signaling and protein folding/sorting in the endoplasmic reticulum. PMID: 23464991
  16. The data presented in this study provide new insights into the role of CLN3 in late endosomal/lysosomal membrane transport. PMID: 22261744
  17. Btn1 controls retrograde sorting by regulating SNARE phosphorylation and assembly, a process that may be adversely affected in Batten Disease patients. PMID: 21987636
  18. The predominant distribution of CLN3 reporter neurons in visual, limbic, and subcortical motor structures of transgenic mice correlates well with the clinical symptoms of juvenile neuronal ceroid lipofuscinosis. PMID: 20875858
  19. CLN6 and CLN3 mutations trigger distinct processes that converge on a shared pathway, responsible for proper subunit c protein turnover and neuronal cell survival. PMID: 21359198
  20. Data suggests that dysfunction of CLN3P may be causative to the disruption of calcium-mediated pathways. PMID: 20933060
  21. Previous reports of genotype and clinical juvenile neuronal ceroid lipofuscinosis phenotype differences were not supported in this investigation, finding no differences between individuals homozygous or heterozygous for the CLN3 deletion. PMID: 20187884
  22. Cln3 is present during crucial periods of neuronal cell differentiation and brain development. PMID: 10509355
  23. Identification of a transactivation motif in the CLN3 protein. PMID: 11699874
  24. Juvenile and variant late infantile neuronal ceroid lipofuscinoses involve mutated CLN genes encoding lysosomal proteins (review). PMID: 12125809
  25. Deletion of glycosylation sites and mutations within conserved amino acid stretches result in slower cell growth and apoptosis. PMID: 12189165
  26. This protein is responsible for Batten Disease. PMID: 12440525
  27. Defective transport at the lysosomal membrane caused by the absence of functional CLN3 is the primary biochemical defect leading to Batten disease. PMID: 14660799
  28. The presence of CLN3 in endosomes of neurons is functionally significant. Endosomal association of the protein was independent of the two lysosomal targeting motifs. PMID: 14699076
  29. The major mutation is a 1.02 kb deletion, removing exons 7 and 8. Both homozygous and heterozygous deletions at the CLN3 gene site have been linked to the clinical syndromes of juvenile neuronal ceroid-lipofuscinosis. PMID: 15032383
  30. These studies identify a novel CLN3 domain that may dictate the localization and function of CLN3. PMID: 15240864
  31. The second cytoplasmic domain of the CLN3 protein contains a dileucine motif and a cluster of acidic amino acids essential for efficient lysosomal targeting. PMID: 15469932
  32. This research suggests a link between CLN3 function, microtubule cytoskeleton, and endocytic membrane trafficking. PMID: 15471887
  33. AP-1 and AP-3 facilitate lysosomal targeting of Batten disease protein CLN3 through its dileucine motif. PMID: 15598649
  34. Batten disease, an inherited neurodegenerative storage disease affecting children, arises from the autosomal recessive inheritance of mutations in Cln3. The protein is located in the lysosomal/endosomal membrane. PMID: 15657902
  35. This study reports a novel mutation identified as a G to T transversion at nucleotide 49 (G49T) in exon 2 of CLN3, introducing a premature stop codon (E17X) near the N-terminus. This represents the most 5' mutation described to date. PMID: 16087292
  36. CLN3 defect in juvenile Batten disease may affect how intracellular levels of arginine are regulated or distributed within the cell. PMID: 16251196
  37. The CLN3 protein trafficked to the vacuole membrane through early endocytic and pre-vacuolar compartments. PMID: 16291725
  38. CLN3P significantly increased the survival rate of SH-SY5Y neuroblastoma cells, further supporting its anti-apoptotic properties. PMID: 16515873
  39. Autophagy is disrupted in juvenile neuronal ceroid lipofuscinosis, likely at the level of autophagic vacuolar maturation, as observed in CLN3 knockin mice. PMID: 16714284
  40. Cooperative signal structures within different cytoplasmic domains of CLN3 are required for efficient sorting and transport to the lysosome. PMID: 17286803
  41. There is a strong correlation between CLN3 protein expression and the synthesis of bis(monoacylglycerol)phosphate. PMID: 17482562
  42. This review provides a concise overview and update on recent research in juvenile neuronal ceroid-lipofuscinosis, specifically focusing on the function of the CLN3 protein. Its primary function may be that of the enzyme palmitoyl-protein delta-9 desaturase. PMID: 17896996
  43. This study concludes that the common mutant CLN3 protein retains significant function, and that juvenile neuronal ceroid lipofuscinoses is a mutation-specific disease phenotype. PMID: 17947292
  44. Homozygous Cln3(delta ex7/8) transgenic mice represent the most suitable disease model for studying the development of the pathogenetic events of juvenile neuronal ceroidlipofuscinoses. PMID: 18265413
  45. CLN3p impacts galactosylceramide transport, raft morphology, and lipid content. PMID: 18317235
  46. A substantial decrease in the transcript level of the truncated CLN3 gene product is observed in cells from 1 kb deletion patients. PMID: 18678598
  47. CLN3 is essential for trafficking along the pathway required for the delivery of lysosomal enzymes. Its loss contributes to and may explain the lysosomal dysfunction underlying Batten disease. PMID: 18817525
  48. CLN3 interacts with Notch and Jun N-terminal kinase signaling pathways. PMID: 19028667
  49. S. pombe btn1, the orthologue of the Batten disease gene CLN3, is required for vacuole protein sorting of Cpy1p and Golgi exit of Vps10p. PMID: 19299465
  50. This study reports a new c.597C>A transversion in exon 8 of the CLN3 gene, homozygous in all affected family members and absent in 200 alleles of normal controls. PMID: 19489875

Show More

Hide All

Database Links

HGNC: 2074

OMIM: 204200

KEGG: hsa:1201

STRING: 9606.ENSP00000353073

UniGene: Hs.534667

Involvement In Disease
Ceroid lipofuscinosis, neuronal, 3 (CLN3)
Protein Families
Battenin family
Subcellular Location
Lysosome membrane; Multi-pass membrane protein. Late endosome. Lysosome. Golgi apparatus. Golgi apparatus membrane. Golgi apparatus, Golgi stack. Golgi apparatus, trans-Golgi network. Cell membrane. Recycling endosome. Membrane raft. Membrane, caveola. Early endosome membrane. Cell junction, synapse, synaptosome. Late endosome membrane. Cytoplasmic vesicle, autophagosome.
Tissue Specificity
Expressed in the cortical brain, pancreas, spleen, and testis with weaker expression in the peripheral nerve (at protein level). Highly expressed in gray matter (at protein level).

Q&A

What is the CLN3 protein and why are reliable antibodies important for its study?

CLN3 (battenin) is a transmembrane protein primarily located in lysosomes and endosomes. Mutations in the CLN3 gene cause juvenile Batten disease (JNCL), characterized by progressive neurodegeneration . Despite its clinical significance, the precise function of CLN3 remains unclear, making specific and reliable antibodies crucial for:

  • Determining protein expression levels in different tissues

  • Identifying subcellular localization

  • Understanding protein-protein interactions

  • Validating disease models

  • Confirming genetic manipulations

The uncertainty surrounding CLN3's function makes high-quality antibodies particularly important, as they represent one of the few tools available to directly study the protein in biological samples .

What issues have been identified with currently available CLN3 antibodies?

Multiple studies have revealed significant specificity problems with both commercial and researcher-generated anti-CLN3 antibodies:

  • Anti-CLN3 antibodies detect the same protein bands in both wildtype and Cln3-/- (knockout) mouse samples

  • These non-specific signals appear consistent across different mouse tissues (cerebellum, cortex, kidney, fibroblasts)

  • Many antibodies fail to specifically detect human CLN3 protein expressed at high levels in laboratory cell lines

  • Cross-reactivity with unrelated proteins generates signals that could be misinterpreted as CLN3

What antibody validation methods should be implemented when working with CLN3 antibodies?

Given the documented specificity issues, researchers should implement rigorous validation protocols:

Required validation steps:

  • Knockout controls: Compare antibody signal between wildtype and Cln3-/- samples to identify non-specific binding

  • Overexpression controls: Test antibody against cells overexpressing tagged CLN3 (e.g., myc-CLN3)

  • Multiple antibody comparison: Use antibodies targeting different CLN3 epitopes and compare results

  • Multiple techniques: Combine immunoblotting with immunofluorescence and other methods

  • Multiple tissue/sample types: Validate across different biological contexts

Sample processing optimization:

  • Test multiple protein extraction conditions (varying detergents, salt concentrations)

  • Evaluate different sample denaturation protocols (37°C for 30 min, 65°C for 15 min, 100°C for 10 min)

  • Include chaotropic agents like urea (4M) to prevent aggregation of hydrophobic membrane proteins

What are the key characteristics of CLN3 antibodies used in published research?

Published studies have utilized various anti-CLN3 antibodies with different characteristics:

SourceAntibody TypeEpitope/ImmunogenPurification MethodSpecies ReactivityImmunoblot DilutionImmunofluorescence Dilution
Anu Jalanko's group (Finland)Rabbit polyclonal m385Synthetic peptide (aa 242-258 of mouse CLN3)Immunogen affinity purifiedMouse, human1:500-1:10001:1000
Tuxworth et al.Rabbit polyclonal 9033Synthetic peptide (aa 5-19 of mouse CLN3)SerumMouse1:50-1:5001:100
Commercial (Abnova)Rabbit polyclonalFull-length human CLN3Not specifiedHuman, mouse1:500-1:1000Not specified
Commercial (Abnova)Mouse monoclonalFull-length human CLN3 with GST tagNot specifiedHuman, mouse1:200-1:1000Not specified
Commercial (Abcam)Rabbit polyclonalSynthetic peptide (aa 400-438 of human CLN3)Not specifiedHuman, mouse1:700Not specified

Despite their diverse characteristics, studies have shown that these antibodies lack sufficient specificity for reliable CLN3 detection .

What alternative approaches can researchers use to study CLN3 when antibody specificity is questionable?

Given the limitations of current CLN3 antibodies, researchers should consider alternative strategies:

  • Epitope tagging: Express CLN3 fusion proteins with reliable tags (myc, FLAG, GFP) that can be detected with well-characterized antibodies

  • Transcript analysis: Use RT-PCR or RNA-seq to quantify CLN3 mRNA expression patterns and variants

  • FRET-based approaches: Utilize fluorescence resonance energy transfer with tagged CLN3 constructs to study protein interactions and topology

  • Genetic models: Create CLN3 knockout/knockin models using CRISPR/Cas9 and analyze phenotypic effects rather than directly detecting the protein

  • Functional assays: Measure relevant lysosomal/endosomal functions as indirect indicators of CLN3 activity

These approaches can provide valuable insights into CLN3 biology while avoiding the pitfalls associated with direct antibody detection .

How can researchers determine if CLN3 antibody signals in Batten disease models represent mutant CLN3 protein or cross-reactivity?

This question addresses a fundamental controversy in the field. Residual staining with CLN3 antibodies has been observed in tissues from homozygous Cln3Δex7/8 mice, but it remains unclear whether this represents:

  • Actual detection of truncated/mutant CLN3 proteins

  • Non-specific binding to unrelated proteins

Methodological approaches to resolve this question:

A. Genetic complementation experiments:

  • Express wild-type CLN3 in patient-derived cells

  • Compare siRNA knockdown effects in patient cells (if signals decrease with siRNA, they likely represent mutant CLN3)

B. Mass spectrometry analysis:

  • Immunoprecipitate with anti-CLN3 antibodies from both wild-type and mutant samples

  • Perform LC-MS/MS to identify all captured proteins

  • Compare protein profiles to identify cross-reactive proteins

C. Correlative microscopy:

  • Combine electron microscopy with super-resolution immunofluorescence

  • Precisely localize antibody signals relative to subcellular structures

  • Compare localization patterns between wild-type and mutant samples

D. mRNA-protein correlation:

  • Quantify mutant CLN3 mRNA expression levels

  • Compare with antibody signal intensity

  • Consistent correlations would support detection of mutant protein

Understanding whether mutant CLN3 proteins retain partial function is critical for developing therapeutic strategies focused on upregulating residual function versus replacing lost function entirely .

What methodological approaches can improve specificity when generating new CLN3 antibodies?

Developing more specific CLN3 antibodies requires strategic approaches to immunogen design and antibody validation:

Immunogen design strategies:

  • Topologically informed epitope selection: Target unique, exposed regions of CLN3 based on membrane topology models (e.g., six transmembrane domains with cytosolic N/C termini)

  • Comparative sequence analysis: Design peptide immunogens from regions that:

    • Are highly conserved across species but unique to CLN3

    • Avoid regions with similarity to other proteins

    • Include residues from transmembrane boundaries rather than hydrophobic cores

  • Conformational epitopes: Generate antibodies against folded domains rather than linear peptides using:

    • Bacterial expression of soluble CLN3 domains

    • Reconstitution of CLN3 in nanodiscs or liposomes

Advanced validation protocols:

  • Multi-step screening:

    • Pre-adsorb antibodies against knockout tissue lysates

    • Screen against arrays of CLN3 peptides/domains

    • Test on samples from multiple species

  • Epitope mapping:

    • Use systematic mutagenesis of key residues

    • Create chimeric proteins between CLN3 and related proteins

  • Cross-validation:

    • Compare results using orthogonal detection methods

    • Validate with techniques like proximity ligation assays

These methodological improvements could significantly advance the field by providing more reliable tools for CLN3 research .

How can researchers investigate CLN3 localization when antibody specificity is questionable?

Given the documented issues with CLN3 antibodies, alternative approaches to study subcellular localization include:

A. Multi-tag transposomic approach:

  • Generate a library of CLN3 cDNA clones with randomly inserted tags (eGFP, myc)

  • Evaluate functionality by assessing ability to rescue phenotypes in CLN3-deficient cells

  • Use double-tagged clones to perform FRET analysis for determining membrane orientation

B. Correlative protein localization:

  • Express CLN3 with split fluorescent/luminescent reporters

  • Combine with established markers for different subcellular compartments

  • Use live-cell imaging to track dynamic localization

C. Proximity labeling approaches:

  • Express CLN3 fused to enzymes like BioID or APEX2

  • Identify proximal proteins through biotinylation

  • Map subcellular environment of CLN3 indirectly

D. Functional domain mapping:

  • Create systematic domain deletions/mutations

  • Identify regions required for proper localization

  • Correlate with known trafficking motifs

These approaches rely less on direct antibody detection and can provide more reliable insights into CLN3 localization and membrane topology . The FRET-assisted approach has already yielded a refined model of CLN3 membrane topology with six transmembrane domains and cytosolic N- and C-termini .

How can researchers reconcile contradictory data from CLN3 antibody studies in the literature?

The CLN3 research field contains seemingly contradictory data regarding protein localization and function, likely due to antibody specificity issues. Researchers can address these contradictions through:

Systematic reanalysis approach:

  • Catalog contradictions: Systematically identify conflicting findings across studies

  • Evaluate antibody characteristics: Compare specificity validation methods used

  • Assess complementary evidence: Identify studies with non-antibody supporting data

  • Replicate key experiments: Use multiple antibodies and controls

  • Validate with genetic approaches: Confirm findings in knockout/rescue systems

Common sources of contradictions to evaluate:

  • Different antibody epitopes and purification methods

  • Variations in sample preparation (detergents, fixation)

  • Cell-type specific differences in CLN3 expression or modification

  • Alternative splice variants of CLN3

  • Differences between overexpressed and endogenous protein

The variable phenotypes observed across different CLN3 disease models may reflect true biological variation rather than experimental artifacts . Careful consideration of alternative CLN3 transcript expression and the potential for residual function in different disease-causing mutations is essential when interpreting contradictory findings .

How can CLN3 antibodies be used to investigate protein-protein interactions despite specificity concerns?

Despite specificity limitations, modified approaches can still yield valuable information about CLN3 protein interactions:

Comparative interaction profiling:

  • Parallel analysis in wildtype and knockout systems:

    • Perform immunoprecipitation with anti-CLN3 antibodies in both systems

    • Identify proteins that associate only in wildtype samples

    • Filter out common interactors as likely non-specific

  • Proximity-dependent approaches:

    • Express CLN3-BioID fusion proteins

    • Compare biotinylated protein profiles between wildtype and mutant CLN3 variants

    • Identify differential interactions that correlate with function

Validated CLN3 interactions:
CLN3 has been shown to interact with several proteins, including:

  • Rab7A (confirmed by BRET and co-immunoprecipitation)

  • Sortilin (confirmed by BRET)

  • Rab1a (weaker interaction confirmed by BRET)

Disease-causing mutations affect these interactions differently:

  • R334H and V330F mutations strengthen Rab7A binding

  • E295K has minimal effect on Rab7A binding

  • L101P disrupts Rab7A binding in co-IP experiments

Understanding these differential effects on protein interactions provides insight into potential disease mechanisms and therapeutic targets, even when using antibodies with suboptimal specificity .

What experimental designs can best utilize CLN3 antibodies in disease model validation?

Despite specificity concerns, strategic experimental designs can leverage CLN3 antibodies for disease model validation:

Comparative phenotypic profiling:

  • Generate parallel disease models (patient-derived cells, knockout cells, animal models)

  • Perform side-by-side analysis of:

    • Lysosomal storage (LC3-II accumulation)

    • Endosomal trafficking markers (sortilin, CI-MPR degradation)

    • Autophagosome-lysosome fusion

  • Compare phenotypes with those observed in patient samples

Functional rescue approach:

  • Express wild-type CLN3 versus disease-causing mutants in knockout systems

  • Measure rescue of characteristic phenotypes:

    • LC3-II accumulation

    • Lysosomal/endosomal receptor degradation

    • Membrane protein trafficking

  • Use antibodies against affected pathways rather than directly against CLN3

These approaches leverage the phenotypic manifestations of CLN3 deficiency rather than relying solely on direct CLN3 detection . Studies have shown that CLN3-deficient cells accumulate LC3II-positive autophagosomes and show accelerated degradation of lysosomal sorting receptors like sortilin and CI-MPR, providing reliable phenotypic markers .

What technologies are emerging to overcome the limitations of traditional CLN3 antibodies?

Several cutting-edge technologies show promise for improving CLN3 research:

Advanced antibody engineering:

  • Single-domain antibodies (nanobodies) with improved specificity

  • Synthetic antibody libraries screened against native membrane proteins

  • Recombinant antibodies with defined binding sites

Alternative protein detection platforms:

  • DNA-barcoded antibody reporting for higher specificity

  • Aptamer-based detection systems

  • Mass cytometry for multi-parameter single-cell analysis

Genetic tagging technologies:

  • Endogenous tagging using CRISPR/Cas9 knock-in

  • Split fluorescent protein complementation

  • Self-labeling protein tags (SNAP, CLIP, Halo) for live-cell imaging

Functional readout systems:

  • Reporter cell lines for CLN3-dependent functions

  • High-content imaging of lysosomal/endosomal trafficking

  • Metabolic profiling of CLN3-dependent pathways

These emerging technologies could significantly advance CLN3 research by providing more reliable detection methods and functional readouts .

How can researchers determine if their CLN3 antibody is suitable for a specific application?

Researchers should implement a systematic validation workflow before using CLN3 antibodies for specific applications:

Application-specific validation protocol:

  • Western blotting validation:

    • Test on multiple positive controls (tissues known to express CLN3)

    • Include negative controls (CLN3 knockout/knockdown samples)

    • Verify expected molecular weight (~48 kDa for full-length CLN3)

    • Optimize protein extraction and sample preparation (detergents, urea)

    • Validate across different sample processing conditions

  • Immunofluorescence validation:

    • Compare staining patterns between wildtype and knockout cells

    • Co-localize with established organelle markers

    • Compare with known CLN3 distribution patterns

    • Test fixation and permeabilization methods

  • Immunoprecipitation validation:

    • Confirm enrichment of CLN3 in IP versus input

    • Perform reciprocal IPs with known interactors

    • Validate interactions by orthogonal methods

  • Flow cytometry validation:

    • Compare signal distributions between positive and negative populations

    • Include isotype controls

    • Validate with cells expressing tagged CLN3

By implementing these rigorous validation protocols for each specific application, researchers can better determine whether their CLN3 antibody is suitable for a particular experimental purpose .

How does the membrane topology of CLN3 affect antibody selection and experimental design?

Understanding CLN3's membrane topology is crucial for effective antibody selection and experimental design:

Current topology model:

  • Six transmembrane domains

  • Cytosolic N- and C-termini

  • Three luminal loops and two cytosolic loops

Topology-informed antibody selection:

  • Epitope accessibility:

    • For intact cell applications, target extracellular/luminal domains

    • For permeabilized cells/lysates, consider all domains

    • Avoid transmembrane regions for antibody generation

  • Disease-mutation considerations:

    • Common 1.02-kb deletion removes transmembrane domains 2-4 and luminal loop 2

    • Missense mutations cluster on the luminal side of the transmembrane structure

    • Antibodies targeting regions affected by mutations may show differential binding

  • Domain-specific applications:

    • Target N/C termini for pull-down experiments

    • Use conformation-sensitive antibodies for functional studies

    • Consider domain-specific antibodies for mapping protein interactions

The FRET-assisted determination of CLN3 topology provides a structural framework for more rational antibody design and application, potentially improving specificity and reliability in future studies .

What are the implications of CLN3 antibody limitations for translational research and therapeutic development?

The limitations of current CLN3 antibodies have significant implications for translational research:

Impact on biomarker development:

  • Uncertain reliability for measuring treatment effects

  • Challenges in quantifying CLN3 protein levels in clinical samples

  • Need for alternative biomarkers of CLN3 function

Therapeutic development considerations:

  • Gene therapy approaches:

    • Need reliable readouts of therapeutic gene expression

    • Challenges in confirming protein production and localization

    • Requirement for functional assays rather than direct protein detection

  • Small molecule development:

    • Difficulty in high-throughput screening using antibody-based assays

    • Reliance on phenotypic screens rather than target-based approaches

    • Need for surrogate markers of CLN3 function

  • Clinical trial design:

    • Limited options for pharmacodynamic biomarkers

    • Challenges in patient stratification based on protein expression

    • Necessity for functional/clinical endpoints

Despite these challenges, understanding the limitations of current tools is essential for planning robust translational research strategies and developing meaningful clinical endpoints for therapeutic trials .

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.