DNAJB6 antibodies target the DnaJ (Hsp40) homolog, subfamily B, member 6 protein, which exists in two splice isoforms:
DNAJB6a: 38 kDa, nuclear-localized isoform.
DNAJB6b: 27 kDa, cytoplasmic isoform with distinct aggregation-suppression activity .
DNAJB6 antibodies are pivotal in studying the protein’s role in:
α-Synuclein Aggregation: DNAJB6b (27 kDa) suppresses α-synuclein aggregation in Parkinson’s disease (PD) models. Knockout (KO) of DNAJB6 increases α-synuclein aggregates by 15% in HEK293T cells .
Lewy Body Pathology: DNAJB6 is enriched in Lewy bodies (LBs) of PD patients, where it interacts with Hsp70 to maintain α-synuclein solubility .
Metastasis Suppression: DNAJB6a (38 kDa) inhibits epithelial-mesenchymal transition (EMT) in breast cancer by upregulating DKK1 (Wnt inhibitor) and downregulating Vimentin and Twist1 .
Clinical Correlation: Reduced DNAJB6a levels correlate with advanced-stage infiltrating ductal carcinoma .
HCMV Replication: DNAJB6 isoforms modulate nuclear import of HCMV UL70 primase, affecting viral DNA synthesis .
Dengue Virus (DENV): DNAJB6 and Hsp70 regulate DENV virion assembly by maintaining viral proteostasis .
LGMD1D Myopathy: DNAJB6 mutants (e.g., F93L, F100I) retain polyglutamine aggregation suppression but exhibit structural instability, leading to toxic gain-of-function in limb-girdle muscular dystrophy .
Wnt/β-Catenin Pathway: DNAJB6a recruits PP2A to activate GSK3β, promoting β-catenin degradation and suppressing oncogenic signaling (Figure 1) .
Proteintech 66587-1-Ig: Mouse monoclonal antibody validated for WB, IHC, and IF .
Proteintech 11707-1-AP: Rabbit polyclonal antibody with IP compatibility and extensive publication records .
Cusabio CSB-PA03239A0Rb: Cost-effective option for WB and IF, available in HRP/FITC conjugates .
DNAJB6 antibodies are versatile research tools with multiple applications in molecular and cellular biology. The primary applications include:
Western Blotting (WB): Detects DNAJB6 protein in cell or tissue lysates with high specificity. Most DNAJB6 antibodies show reactivity in WB applications with recommended dilutions typically ranging from 1:2000 to 1:8000 .
Immunoprecipitation (IP): Isolates DNAJB6 protein complexes from cell lysates to study protein-protein interactions, particularly useful when examining DNAJB6's interactions with other chaperone proteins .
Immunofluorescence (IF): Visualizes subcellular localization of DNAJB6 in fixed cells, enabling distinction between nuclear DNAJB6a and cytoplasmic DNAJB6b isoforms .
Immunohistochemistry (IHC): Detects DNAJB6 expression in tissue sections, with most antibodies requiring antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) .
Enzyme-linked Immunosorbent Assay (ELISA): Quantifies DNAJB6 levels in various biological samples .
Flow Cytometry (FACS): Analyzes DNAJB6 expression in individual cells within heterogeneous cell populations .
Multiple validated DNAJB6 antibodies are available with reactivity to human, mouse, and rat proteins, making them suitable for diverse experimental models .
Selecting the optimal DNAJB6 antibody requires careful consideration of several key factors:
1. Isoform specificity:
DNAJB6 exists in two main spliced isoforms (DNAJB6a and DNAJB6b) with distinct subcellular localizations and functions
For isoform-specific detection, select antibodies targeting unique regions, such as anti-DNAJB6b antibodies that recognize the CKEQLLRLDNK sequence at amino acids 232-240
General DNAJB6 antibodies typically recognize common epitopes present in both isoforms
2. Application compatibility:
Verify the antibody is validated for your specific application (WB, IP, IF, IHC, ELISA, or FACS)
Some antibodies perform better in certain applications than others; for example, monoclonal DNAJB6 antibodies like B-5 (sc-365574) are validated for WB, IP, IF, and ELISA
3. Species reactivity:
Confirm reactivity with your experimental model (human, mouse, rat, etc.)
Many DNAJB6 antibodies detect conserved epitopes across species
4. Clonality considerations:
Monoclonal antibodies offer high specificity but recognize single epitopes
Polyclonal antibodies provide broader recognition but may have batch-to-batch variation
5. Format and conjugation:
Choose between unconjugated or conjugated antibodies (HRP, PE, FITC, Alexa Fluor®) based on your detection system
For multiplex staining, consider fluorescently conjugated antibodies
Review product validation data, published literature, and supplementary resources to guide your selection decision .
The following protocol outlines the optimal steps for detecting DNAJB6 using Western blotting:
Sample preparation:
Lyse cells or tissues in appropriate lysis buffer (RIPA buffer containing protease inhibitors is commonly used)
Determine protein concentration using standard methods (BCA or Bradford assay)
Prepare samples by mixing with Laemmli buffer and heating at 95°C for 5 minutes
Gel electrophoresis and transfer:
Load 20-40 μg of protein per lane on 10-12% SDS-PAGE gels
Separate proteins at 100-120V until adequate resolution is achieved
Transfer proteins to PVDF or nitrocellulose membrane (wet transfer recommended)
Antibody incubation:
Block membrane with 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Incubate with primary DNAJB6 antibody at recommended dilution (typically 1:2000-1:8000) overnight at 4°C
Wash membrane 3-5 times with TBST, 5 minutes each
Incubate with appropriate HRP-conjugated secondary antibody for 1 hour at room temperature
Wash membrane 3-5 times with TBST, 5 minutes each
Detection:
Apply ECL substrate and detect signal using film or digital imaging systems
Expected molecular weights:
Notes:
For optimal isoform separation, use 10% gels and extend running time
When analyzing DNAJB6 protein half-life, cycloheximide chase assays can be employed with Western blotting as the detection method
For validation, include positive controls (brain tissue extracts show high expression of DNAJB6)
When experiencing issues with DNAJB6 immunostaining, consider the following troubleshooting approaches:
1. High background or non-specific staining:
Increase blocking time or concentration (try 5% normal serum matching the species of secondary antibody)
Reduce primary antibody concentration (test serial dilutions)
Include additional washing steps with 0.025-0.5% Triton X-100 in PBS
Use appropriate controls (secondary antibody only, isotype controls)
2. Weak or no signal:
Optimize antigen retrieval method:
Increase primary antibody concentration or incubation time
Test different fixation methods (4% PFA for 20 minutes is standard for cultured cells)
For cells, include membrane permeabilization step with 0.5% Triton-X-100 for 5 minutes
3. Unexpected staining pattern:
Verify antibody specificity (validate with KO/KD controls)
Consider isoform-specific staining - DNAJB6a is nuclear while DNAJB6b is cytoplasmic
F-actin counterstain (phalloidin-atto550) can help visualize cell boundaries
4. Tissue-specific considerations:
For brain tissue, co-staining with tyrosine hydroxylase (TH) can help identify specific neuronal populations
When staining disease models (e.g., synucleinopathies), consider that DNAJB6b may be downregulated
5. Technical optimization:
For immunofluorescence, minimize exposure to light during secondary antibody incubation
Consider using automated staining platforms for consistent results
Document all optimization steps systematically
For difficult samples, pre-absorption of antibody with antigenic peptide can be used as a specificity control .
Distinguishing between DNAJB6 isoforms requires specific approaches given their high sequence similarity:
Isoform-specific antibodies:
Use isoform-specific antibodies like anti-DNAJB6b antibodies that target unique C-terminal sequences (amino acids 232-240: CKEQLLRLDNK)
Commercial antibodies may recognize both isoforms; verify specificity with recombinant proteins expressing either isoform
For custom antibody generation, target the unique C-terminal regions of each isoform
Immunofluorescence patterns:
DNAJB6a localizes predominantly to the nucleus
DNAJB6b shows cytoplasmic distribution
Counterstain with DAPI for nuclear visualization and F-actin stain (phalloidin-atto550) for cytoplasmic boundaries
Co-localization studies with nuclear and cytoplasmic markers confirm isoform identities
Western blotting differentiation:
DNAJB6a appears at approximately 38 kDa
DNAJB6b appears at approximately 27 kDa
Use 10-12% SDS-PAGE gels with extended running time for optimal separation
Include appropriate positive controls (brain tissue expresses both isoforms)
Functional validation:
DNAJB6a effectively suppresses nuclear polyQ protein aggregation
DNAJB6b is a potent suppressor of cytoplasmic polyQ aggregation
Design experiments testing these differential activities
RT-PCR approach:
Design primers flanking the alternatively spliced regions
Amplification produces distinct band sizes for each isoform
Combine with Western blotting for comprehensive analysis
Implementing these strategies allows precise discrimination between the functionally distinct DNAJB6 isoforms, critical for accurately interpreting research findings .
Multiple complementary techniques can effectively characterize DNAJB6 protein-protein interactions:
Co-immunoprecipitation (Co-IP):
Use DNAJB6 antibodies coupled to agarose beads for pull-down experiments
For higher specificity, use DNAJB6 Antibody AC (sc-365574 AC) containing 25% agarose
After immunoprecipitation, analyze complexes by Western blotting with antibodies against potential interaction partners
Verify interactions bidirectionally by immunoprecipitating with antibodies against the partner protein
Controls should include non-specific IgG and DNAJB6 or partner antibodies alone
Proximity ligation assay (PLA):
Enables visualization of protein interactions in situ with single-molecule sensitivity
Requires specific antibodies raised in different species against DNAJB6 and its potential partners
Particularly useful for detecting interactions between DNAJB6 and other chaperones like Hsp70, BAG3, and HSPB8
Bimolecular fluorescence complementation (BiFC):
Express DNAJB6 and potential interactor fused to complementary fragments of fluorescent proteins
Interaction brings fragments together, restoring fluorescence
Allows visualization of interaction sites within cells
Mass spectrometry-based approaches:
Tandem affinity purification coupled with mass spectrometry identifies novel interaction partners
SILAC or TMT labeling enables quantitative comparison of interactomes between wild-type and mutant DNAJB6
Particularly valuable for studying disease-causing mutations that may alter protein interactions
Yeast two-hybrid screening:
Identifies direct protein-protein interactions
Can be used to screen libraries for novel DNAJB6 interactors
Validate hits with other methods due to potential false positives
In vitro binding assays:
Recombinant DNAJB6 and potential partners can be used in pull-down assays
Surface plasmon resonance (SPR) quantifies binding kinetics and affinities
Especially important for characterizing DNAJB6's interaction with Hsp70 and its role in chaperoning activity
These methodologies have revealed DNAJB6's interactions with key partners in protein quality control pathways, including components of the chaperone-assisted selective autophagy (CASA) complex .
Investigating DNAJB6 mutations requires a comprehensive approach combining multiple antibody-based techniques:
Protein stability and turnover:
Cycloheximide chase assay: Transfect cells with wild-type or mutant DNAJB6, block protein synthesis with cycloheximide, and quantify remaining protein over time using Western blotting
Research shows mutations p.Phe93Leu and p.Phe89Ile significantly decrease DNAJB6 turnover rates
Complement with proteasome inhibitors (lactacystin) or lysosome inhibitors (bafilomycin A1) to determine degradation pathways
Subcellular localization:
Immunofluorescence microscopy to detect altered localization patterns of mutant DNAJB6
Co-staining with organelle markers (nuclear, ER, mitochondrial) to identify mislocalization
Live-cell imaging with fluorescently-tagged DNAJB6 variants to track dynamic localization changes
Protein aggregation:
Filter trap assays with anti-DNAJB6 antibodies to detect SDS-insoluble aggregates
Immunofluorescence to visualize aggregation patterns and co-localization with other proteins
Sequential extraction protocols to assess solubility changes in mutant DNAJB6
Chaperone activity:
Luciferase refolding assays with immunoprecipitation of DNAJB6 complexes
Polyglutamine aggregation suppression assays (nuclear vs. cytoplasmic) to evaluate isoform-specific functions
Co-immunoprecipitation to assess Hsp70 binding efficiency of mutant DNAJB6
Autophagy dysregulation:
Immunoblotting for autophagy markers (LC3-II, p62/SQSTM1) in cells expressing mutant DNAJB6
Immunofluorescence co-localization studies with autophagy markers
Analysis of CASA complex formation using proximity ligation assays
Disease model analysis:
Immunohistochemistry in patient samples or animal models with DNAJB6 mutations
DNAJB6 expression levels by quantitative ELISA in clinical specimens
Mitochondrial dysfunction assessment via co-localization with mitochondrial markers
These methodologies have revealed that DNAJB6 mutations associated with limb-girdle muscular dystrophy (LGMD D1) cause protein accumulation and disrupt protein homeostasis, potentially contributing to mitochondrial abnormalities and muscle weakness .
Investigating DNAJB6 in neurodegenerative disease contexts requires specialized approaches:
Tissue-specific considerations:
Brain tissue requires optimized fixation and antigen retrieval protocols:
Use co-staining with neuronal markers (e.g., tyrosine hydroxylase) to identify specific populations
Pathological protein aggregates:
DNAJB6 is present in Lewy bodies in Parkinson's disease patients
Use dual immunofluorescence to co-localize DNAJB6 with α-synuclein, tau, or polyQ proteins
Apply sequential extraction protocols to evaluate DNAJB6 in soluble versus insoluble fractions
Isoform-specific alterations:
Use isoform-specific antibodies for precise quantification
Apply quantitative ELISA methods developed specifically for DNAJB6 detection
Protein-protein interactions in disease:
Investigate DNAJB6 interaction with:
Use proximity ligation assays in tissue sections for in situ interaction detection
Functional rescue experiments:
Overexpress wild-type DNAJB6 in disease models and assess:
Autophagy pathway involvement:
Monitor DNAJB6's interaction with chaperone-assisted selective autophagy (CASA) components
Assess co-localization with LC3-positive autophagosomes
Evaluate impact of autophagy modulators (e.g., LiCl, which may work through GSK3β inhibition and autophagy activation)
These approaches have revealed that DNAJB6 plays protective roles in neurodegenerative diseases by preventing protein aggregation and promoting clearance of misfolded proteins .
Comprehensive control strategies ensure reliable and interpretable results when using DNAJB6 antibodies:
Positive controls:
Tissues/cells known to express DNAJB6:
Recombinant DNAJB6 protein for Western blotting standardization
DNAJB6-overexpressing cells as technical positive controls
Negative controls:
Primary antibody omission (secondary antibody only)
Isotype control antibodies matching the DNAJB6 antibody host species and class
DNAJB6 knockdown/knockout cells or tissues (validated by genotyping)
Non-expressing tissues (use tissue panels to identify suitable negative controls)
Specificity controls:
Pre-absorption with immunizing peptide should abolish specific signal
Comparison of staining patterns between different DNAJB6 antibodies targeting distinct epitopes
Western blotting to confirm single bands at expected molecular weights (27 kDa for DNAJB6b, 38 kDa for DNAJB6a)
Isoform validation:
Compare antibodies targeting common regions versus isoform-specific regions
Subcellular localization patterns (nuclear for DNAJB6a, cytoplasmic for DNAJB6b)
Simultaneous detection with isoform-specific primers in RT-PCR
Application-specific controls:
For immunoprecipitation: IgG control and DNAJB6 antibody alone
For immunofluorescence: Counterstains to verify subcellular localization (DAPI for nucleus, phalloidin for cytoskeleton)
For FACS: Single-stained and unstained populations
Disease model controls:
Age-matched and gender-matched controls for patient samples
Wild-type littermates for transgenic animal models
Vehicle-treated samples for drug intervention studies
Rigorous implementation of these controls ensures that observed signals represent genuine DNAJB6 detection rather than technical artifacts or non-specific binding.
Multiple complementary approaches enable accurate quantification of DNAJB6 expression:
Western blotting quantification:
Use standard curves with recombinant DNAJB6 protein
Normalize DNAJB6 signal to housekeeping proteins (β-actin, GAPDH, or α-tubulin)
Employ image analysis software for densitometry (ImageJ, Image Lab)
For isoform-specific quantification, distinguish 27 kDa (DNAJB6b) from 38 kDa (DNAJB6a) bands
Include multiple technical and biological replicates (n≥3)
DNAJB6-specific ELISA:
Novel quantitative ELISA methods developed specifically for DNAJB6 detection provide greater sensitivity
Establish standard curves using purified recombinant DNAJB6
For isoform-specific quantification, use capture or detection antibodies targeting unique epitopes
Validate results against Western blotting measurements
Immunohistochemistry/Immunofluorescence quantification:
Digital image analysis of stained tissue sections
Measure:
Staining intensity (mean fluorescence intensity)
Percentage of positive cells
Subcellular distribution patterns
Use automated tissue analysis platforms for consistent results
Consider regional variations in expression, particularly in brain tissue
Flow cytometry:
Quantify DNAJB6 expression in single cells within heterogeneous populations
Establish gating strategies based on negative controls
Calculate mean fluorescence intensity (MFI) for population-level comparisons
Particularly useful for blood cells or disaggregated tissue samples
qRT-PCR for mRNA quantification:
Design primers specific to DNAJB6 (for total expression) or spanning unique regions (for isoform-specific detection)
Normalize to stable reference genes
Correlate mRNA levels with protein expression to assess post-transcriptional regulation
Note that protein levels may not directly correlate with mRNA levels due to post-transcriptional regulation
Mass spectrometry-based proteomics:
Selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) for absolute quantification
Use stable isotope-labeled peptide standards for precise measurement
Can distinguish isoforms based on unique peptide sequences
These methods have revealed differential expression patterns of DNAJB6 in disease states, such as downregulation of DNAJB6b in synucleinopathies and altered expression in cancer tissues .
Creating custom DNAJB6 antibodies requires careful planning and comprehensive validation:
Antigen design strategies:
For pan-DNAJB6 antibodies:
Target conserved regions between isoforms, typically within amino acids 1-217
The J-domain (amino acids ~1-70) is highly conserved among DnaJ proteins and may cause cross-reactivity
For isoform-specific antibodies:
Avoid regions with post-translational modifications that might mask epitopes
Immunization considerations:
Conjugate peptides to carrier proteins (KLH, BSA) to enhance immunogenicity
For DNAJB6b-specific antibodies, include an N-terminal cysteine for conjugation
Use multiple immunization protocols with varied schedules (5 doses over 12 weeks shows good results)
Consider host species that differ from experimental models to minimize background
Purification strategies:
Two-step purification process:
Elute bound antibodies with glycine pH 2.7 into Tris-containing vials
Validation requirements:
Western blotting: Confirm single bands at expected molecular weights (27 kDa for DNAJB6b, 38 kDa for DNAJB6a)
Immunoprecipitation: Verify ability to pull down endogenous DNAJB6
Immunofluorescence: Confirm expected subcellular localization patterns
Knockout/knockdown controls: Test antibody specificity in DNAJB6-depleted samples
Cross-reactivity testing: Evaluate against related DNAJ family proteins
Peptide competition assays: Pre-incubation with immunizing peptide should abolish signal
Documentation requirements:
Detailed information on immunogen sequence and position
Host species and clonality
Validation data across multiple applications
Recommended storage conditions and working dilutions
Species reactivity information
Quality control measures:
Batch-to-batch consistency testing
Stability testing under various storage conditions
Application-specific optimization
Detailed documentation of validation experiments
Successful custom antibody generation has enabled critical discoveries, such as the development of DNAJB6b-specific antibodies that demonstrated downregulation of this isoform in synucleinopathies .
Optimal fixation and sample preparation protocols significantly impact DNAJB6 detection quality:
Fixation considerations:
For cell cultures:
For tissue sections:
4% PFA perfusion followed by post-fixation provides optimal results
Fixation time should be optimized based on tissue thickness (typically 24-48 hours)
Cryopreservation after fixation maintains many epitopes better than paraffin embedding
Antigen retrieval requirements:
For paraffin-embedded tissues:
For frozen sections:
Brief fixation in cold acetone may be sufficient
Mild antigen retrieval may still improve signal intensity
Permeabilization protocols:
For cell cultures:
For tissue sections:
Permeabilization time may need extension for dense tissues
Detergent concentration should be optimized based on tissue type
Blocking conditions:
5% normal serum matching the species of secondary antibody reduces background
For highly autofluorescent tissues (brain, liver), include additional blocking steps:
Sudan Black B treatment
Autofluorescence quenching reagents
Photobleaching prior to antibody incubation
Special considerations for subcellular compartments:
Nuclear DNAJB6a detection may require stronger permeabilization
Cytoplasmic DNAJB6b detection benefits from counterstaining with F-actin (phalloidin-atto550)
Mitochondrial co-localization studies require careful membrane preservation
Tissue-specific adaptations:
Brain tissue: More aggressive antigen retrieval may be needed
Muscle tissue: Extended fixation time can cause overfixation and epitope masking
Clinical specimens: Standardize processing time to minimize pre-analytical variables
These optimized protocols have been critical for discoveries such as the identification of DNAJB6 in Lewy bodies in Parkinson's disease patients and the characterization of DNAJB6 expression patterns in various cell types.
DNAJB6 antibodies serve as powerful tools for investigating neurodegenerative disorders through multiple approaches:
Parkinson's disease research:
Immunohistochemistry reveals DNAJB6 localization in Lewy bodies
Co-immunostaining with α-synuclein antibodies demonstrates direct association
Proximity ligation assays detect interactions between DNAJB6 and parkinsonian proteins
Western blotting shows DNAJB6b downregulation in synucleinopathies
Use isoform-specific antibodies to track differential regulation of DNAJB6a versus DNAJB6b
Huntington's disease applications:
Filter trap assays with anti-DNAJB6 antibodies assess polyQ protein aggregation suppression
DNAJB6's C-terminal domain (amino acids 152-232) is crucial for suppressing polyQ aggregation
Immunofluorescence distinguishes between DNAJB6a's role in nuclear versus DNAJB6b's role in cytoplasmic polyQ protein aggregation
Co-immunoprecipitation reveals polyQ protein interactions with DNAJB6
Protein clearance mechanisms:
Immunofluorescence tracks DNAJB6's role in mitophagy by promoting relocation of Parkin and LC3 to depolarized mitochondria
Western blotting for autophagy markers (LC3-II, p62/SQSTM1) in relation to DNAJB6 levels
Co-immunoprecipitation identifies interactions with chaperone-assisted selective autophagy (CASA) components
Stress response visualization:
Stress granule co-localization studies using immunofluorescence
Heat shock response experiments tracking DNAJB6 relocalization
RNA-binding protein (TDP-43, hnRNPA1, hnRNPA2) co-localization with DNAJB6 under stress conditions
Therapeutic intervention assessment:
Western blotting quantifies DNAJB6 upregulation in response to potential therapeutics
Immunohistochemistry evaluates changes in aggregation patterns following treatment
ELISA measures DNAJB6 levels in patient fluids as potential biomarkers
LiCl treatment effects can be monitored through DNAJB6 antibody-based techniques
These methods have revealed critical roles for DNAJB6 in neurodegenerative diseases, including its presence in the core of Lewy bodies, upregulation in astrocytes of Parkinson's disease patients, and differential isoform activities in suppressing protein aggregation .
DNAJB6 antibodies are essential tools for investigating its critical role in limb-girdle muscular dystrophy (LGMD D1):
Pathological assessment:
Immunohistochemistry identifies characteristic pathological features in muscle biopsies:
Rimmed vacuoles
Protein aggregates
Abnormal fiber morphology
Co-immunostaining with sarcomeric proteins (desmin, α-actinin, FHL1) reveals their accumulation in aggregates
Z-disc protein co-localization studies help characterize structural abnormalities
Mutation impact analysis:
Western blotting quantifies mutant DNAJB6 protein stability:
Immunoprecipitation assesses how mutations affect interactions with other proteins
Filter trap assays measure aggregation-prone protein accumulation
Mitochondrial dysfunction:
Co-immunostaining with mitochondrial markers evaluates structural and functional changes
Loss of DNAJB6 causes mitochondrial defects that contribute to muscle weakness in LGMD D1
Proximity ligation assays detect altered interactions between DNAJB6 and mitochondrial proteins
Therapeutic monitoring:
Western blotting quantifies changes in DNAJB6 levels following treatment
LiCl treatment effects can be tracked through antibody-based methods:
Immunofluorescence assesses changes in protein aggregation patterns
Autophagy pathway investigation:
Co-immunostaining with autophagy markers (LC3, p62/SQSTM1) reveals accumulation in patient muscle
Proximity ligation assays detect DNAJB6 interactions with chaperone-assisted selective autophagy (CASA) components BAG3 and HSPB8
Western blotting quantifies changes in autophagy markers following treatment
Animal model validation:
Immunohistochemistry confirms DNAJB6 mutant mouse models recapitulate human pathology
Western blotting compares protein levels between models and human samples
Developmental studies track DNAJB6 expression during muscle formation and maintenance
These approaches have revealed that dominant mutations in DNAJB6 disrupt protein homeostasis through effects on Hsp70 function, leading to accumulation of misfolded proteins and myopathic changes .
DNAJB6 antibodies enable multifaceted investigation of its emerging roles in cancer:
Differential expression analysis:
Immunohistochemistry on tissue microarrays compares DNAJB6 levels across cancer types and stages
Western blotting quantifies isoform-specific expression changes:
ELISA methods provide quantitative measurement in patient samples
Subcellular localization studies:
Immunofluorescence reveals compartment-specific changes in cancer cells
Nuclear-cytoplasmic distribution of DNAJB6 isoforms may shift during cancer progression
Co-localization with cancer-associated proteins identifies potential functional interactions
Functional mechanism investigation:
Co-immunoprecipitation identifies cancer-specific protein interaction partners
Chromatin immunoprecipitation (ChIP) assesses DNAJB6a's role in transcriptional regulation
Proximity ligation assays visualize protein-protein interactions in situ
Analysis of the secreted proteome from DNAJB6a-expressing cells reveals:
Metastasis and invasion studies:
Immunohistochemistry on primary tumors versus metastatic lesions tracks DNAJB6 changes
DNAJB6a overexpression in aggressive breast cancer cell lines decreases migration and invasion
Xenograft model analysis shows DNAJB6a restricts orthotopic tumor growth in nude mice
Co-immunostaining with epithelial-mesenchymal transition markers correlates with invasion capacity
Clinical correlation analysis:
Tissue microarray staining with DNAJB6 antibodies enables correlation with:
Patient survival outcomes
Response to specific therapies
Cancer molecular subtypes
Multi-parameter immunofluorescence combines DNAJB6 with other prognostic markers
Therapeutic response monitoring:
Western blotting tracks DNAJB6 expression changes following treatment
Immunofluorescence assesses subcellular redistribution after drug exposure
Co-immunoprecipitation identifies altered protein interactions in response to therapy
These approaches have revealed that DNAJB6a functions as a metastasis suppressor in breast cancer, with its expression significantly reduced in aggressive breast cancer cells, while therapeutic restoration of DNAJB6a expression may represent a potential treatment strategy .
DNAJB6 antibodies enable detailed investigation of its emerging roles in viral pathogenesis:
Viral infection models:
Immunofluorescence tracks DNAJB6 redistribution during viral infection
Co-localization studies with viral proteins identify direct interactions
Western blotting quantifies expression changes in response to infection
The chaperone-co-chaperone couple, Hsp70 and DNAJB6, play a determinative role in dengue virus (DENV) virion production
Virion assembly analysis:
Immunoprecipitation of DNAJB6 complexes from infected cells identifies associated viral components
Proximity ligation assays visualize DNAJB6-viral protein interactions in situ
Subcellular fractionation followed by Western blotting locates DNAJB6 in relation to viral assembly sites
Analysis of protein assembly processes responsible for maintaining viral proteostasis
Mechanistic investigation:
Cycloheximide chase assays determine if DNAJB6 affects viral protein stability
Co-immunoprecipitation identifies interactions with viral proteins and other host factors
Time-course immunofluorescence tracks DNAJB6 dynamics throughout viral life cycle
Assessment of how viruses highjack the chaperoning activity of DNAJB6
Therapeutic target exploration:
Antibody-based screening identifies compounds that disrupt DNAJB6-viral protein interactions
Western blotting quantifies changes in viral protein levels following modulation of DNAJB6 activity
Immunofluorescence assesses viral replication efficiency in DNAJB6-modulated cells
Co-immunoprecipitation evaluates therapeutic disruption of chaperone complexes
Host response characterization:
Immunohistochemistry on infected tissues maps DNAJB6 distribution
Flow cytometry quantifies DNAJB6 levels in infected versus uninfected cells
Western blotting tracks post-translational modifications of DNAJB6 during infection
Time-course analysis reveals dynamic changes in DNAJB6 expression and localization
Cross-viral comparisons:
Comparative analysis across multiple virus families:
These approaches have revealed that DNAJB6 plays a crucial role in dengue virus propagation, with potential implications for other viral infections, suggesting that DNAJB6-virus interactions may represent promising drug targets for antiviral therapy .
A comprehensive validation strategy ensures reliable DNAJB6 detection across applications:
Specificity validation:
Genetic approach:
Test antibody in DNAJB6 knockout/knockdown models
Compare signal in wild-type versus DNAJB6-deficient samples
Rescue experiments with DNAJB6 re-expression
Peptide competition:
Orthogonal detection:
Verify consistent results using multiple antibodies targeting different DNAJB6 epitopes
Compare monoclonal versus polyclonal antibody performance
Application-specific validation:
Western blotting:
Immunoprecipitation:
Immunohistochemistry/Immunofluorescence:
Quantitative performance metrics:
Sensitivity assessment:
Determine limit of detection using serial dilutions
Compare detection threshold across applications
Dynamic range evaluation:
Test antibody performance across wide concentration ranges
Establish linear detection ranges for quantification
Reproducibility testing:
Inter-lot consistency evaluation
Intra-laboratory and inter-laboratory reproducibility assessment
Stability over time under recommended storage conditions
Multi-parameter validation:
Multiplexing compatibility:
Test performance in multi-color immunofluorescence
Validate with multiple detection systems (fluorescent, chromogenic)
Cross-platform consistency:
Compare results between Western blotting, immunohistochemistry, and ELISA
Reconcile any discrepancies with biological explanations
Documentation standards:
Comprehensive reporting of validation experiments
Inclusion of positive and negative controls in published images
Disclosure of antibody source, catalog number, lot number, and dilution
Detailed methodological description enabling reproduction by other researchers
Implementation of these validation steps ensures reliable, reproducible DNAJB6 antibody performance across diverse experimental conditions.
Effective experimental design for DNAJB6 functional studies requires careful planning:
Isoform-specific considerations:
Design studies to distinguish between DNAJB6a and DNAJB6b functions:
DNAJB6a: Nuclear localization, transcriptional regulation, nuclear protein quality control
DNAJB6b: Cytoplasmic localization, protein aggregation suppression, stress response
Use isoform-specific antibodies or tagged constructs for precise tracking
Include both isoforms in functional analyses when possible
Loss-of-function approaches:
siRNA/shRNA knockdown:
Design targeting sequences common to both isoforms or isoform-specific regions
Validate knockdown by Western blotting and qRT-PCR
Include scrambled siRNA controls
CRISPR/Cas9 gene editing:
Dominant-negative mutants:
H31Q mutation disrupts J-domain function
Validate expression by Western blotting and functional impact by chaperone assays
Gain-of-function strategies:
Overexpression systems:
Use inducible promoters for temporal control
Create stable cell lines for consistent expression
Include both wild-type and disease-associated mutant forms
Validate expression levels by Western blotting with DNAJB6 antibodies
Domain-specific constructs:
Disease-relevant models:
Neurodegenerative disease models:
PolyQ protein aggregation assays
α-synuclein aggregation models
Patient-derived iPSCs differentiated into neurons
Muscular dystrophy models:
Protein quality control assays:
Luciferase refolding assays to measure chaperone activity
Filter trap assays for aggregation suppression
Cycloheximide chase experiments for protein stability analysis
Proteasome and autophagy inhibitor studies to determine degradation pathways
Stress response analyses:
Heat shock experiments with temperature and duration optimization
Oxidative stress exposure (H₂O₂, paraquat)
ER stress induction (tunicamycin, thapsigargin)
Time-course immunofluorescence to track DNAJB6 relocalization
Interaction studies:
Co-immunoprecipitation with key partners (Hsp70, BAG3, HSPB8)
Proximity ligation assays for in situ interaction visualization
FRET/BRET approaches for real-time interaction dynamics
Mass spectrometry-based interactome analysis
These design considerations enable comprehensive functional characterization of DNAJB6 across diverse cellular contexts and disease states.
DNAJB6 represents a promising biomarker candidate for neurodegenerative diseases, with antibody-based detection methods enabling clinical translation:
Tissue-based biomarker approaches:
Immunohistochemistry on post-mortem brain tissue:
Quantify DNAJB6 expression patterns across disease states
Assess DNAJB6 localization in protein aggregates (Lewy bodies, polyQ inclusions)
Correlate with disease severity and progression markers
Multi-parameter immunofluorescence:
Combine DNAJB6 with disease-specific markers (α-synuclein, tau, TDP-43)
Create diagnostic algorithms based on co-localization patterns
Digital pathology quantification for objective assessment
Fluid biomarker development:
Quantitative ELISA methods for DNAJB6 detection in:
Cerebrospinal fluid (CSF)
Plasma/serum
Extracellular vesicles
Isoform-specific quantification using antibodies targeting unique epitopes:
Multiplex immunoassays combining DNAJB6 with established neurodegeneration markers
Single-cell analysis approaches:
Flow cytometry on peripheral blood mononuclear cells:
Measure DNAJB6 levels in specific immune cell populations
Correlate with disease state and progression
Mass cytometry (CyTOF) for high-dimensional analysis:
Combine DNAJB6 with multiple cellular markers
Identify disease-specific cellular signatures
Digital biomarker integration:
Correlate DNAJB6 levels with:
Neuroimaging data (MRI, PET)
Clinical scales and progression rates
Genetic risk factors
Other protein biomarkers
Machine learning approaches to identify DNAJB6-based prediction models
Longitudinal monitoring applications:
Serial sampling to track DNAJB6 changes during disease progression
Assessment of treatment response using DNAJB6 as a pharmacodynamic marker
Correlation with clinical outcomes and disease milestones
Validation requirements:
Multi-center cohort studies with standardized protocols
Analytical validation of DNAJB6 detection methods:
Precision, accuracy, reproducibility
Pre-analytical variable assessment
Clinical validation against gold-standard diagnosis
These approaches build on research showing DNAJB6b downregulation in synucleinopathies and DNAJB6's presence in pathological protein aggregates, suggesting its potential as both a diagnostic and prognostic biomarker for neurodegenerative diseases.
Cutting-edge technologies are transforming our ability to investigate DNAJB6's functions and interactions:
Advanced imaging approaches:
Super-resolution microscopy:
STORM/PALM imaging achieves 10-20 nm resolution of DNAJB6 nanoscale organization
SIM reveals DNAJB6 distribution within subcellular structures
Expansion microscopy physically enlarges specimens for enhanced visualization
Live-cell imaging techniques:
FRAP (Fluorescence Recovery After Photobleaching) measures DNAJB6 mobility
Single-molecule tracking follows individual DNAJB6 molecules in real-time
Optogenetic control of DNAJB6 localization and interactions
Proximity-based interaction detection:
BioID/TurboID proximity labeling:
Fuse DNAJB6 to biotin ligase to identify proximal proteins
Map interaction neighborhoods in different cellular compartments
Compare interactomes between wild-type and mutant DNAJB6
APEX2 proximity labeling:
Higher spatial and temporal resolution than BioID
Compatible with electron microscopy visualization
Split-protein complementation assays:
NanoBiT for sensitive detection of protein-protein interactions
SPARK (Specific Protein Association tool giving transcriptional Readout with rapid Kinetics) for detecting transient interactions
Proteomics innovations:
Crosslinking mass spectrometry (XL-MS):
Maps interaction interfaces between DNAJB6 and client proteins
Identifies conformational changes in disease-associated mutations
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Measures structural dynamics and conformational changes
Identifies regions involved in client binding
Thermal proteome profiling:
Assesses DNAJB6 thermal stability changes upon client binding
Identifies small molecules that stabilize or destabilize DNAJB6
Single-cell technologies:
Single-cell proteomics:
Measures DNAJB6 levels and modifications in individual cells
Correlates with cellular phenotypes and states
Spatial transcriptomics combined with immunofluorescence:
Maps DNAJB6 protein expression alongside transcriptome data
Provides spatial context for DNAJB6 function
Structural biology approaches:
Cryo-electron microscopy:
Determines structures of DNAJB6 complexes with client proteins and Hsp70
Visualizes conformational changes during chaperone cycle
Integrative structural biology:
Combines multiple data types (SAXS, NMR, XL-MS, cryo-EM)
Creates comprehensive structural models of DNAJB6 function
Functional genomics integration:
CRISPR screens with DNAJB6 antibody-based readouts
Parallel reporter assays to study DNAJB6 transcriptional effects
Synthetic genetic interaction mapping to identify functional pathways
These emerging technologies provide unprecedented insights into DNAJB6's dynamic functions across cellular contexts and disease states, enabling mechanistic understanding that may guide therapeutic development.