The TBC1D25 antibody is a research tool designed to detect and study the TBC1 domain family member 25 protein, a Rab GTPase-activating protein (GAP) involved in autophagy regulation. This antibody targets TBC1D25 (UniProt ID: Q3MII6), which facilitates autophagosome-lysosome fusion by modulating Rab33B activity .
Immunogen: Derived from a fusion protein containing the sequence AVDPQITSLDVLQHILIRAFDLSGKKNFGISYLGRDRLGQEVYLSLLSDWDLSTAFATASKPYLQLRVDIRPSEDSPLLEDWDIISPKDVSGSDVLLAEKRSSLTTAALPFTQSIL .
Cross-Reactivity: Recognizes human, mouse, and rat orthologs with 95–96% sequence identity .
Molecular Weight: Detects a band at ~80 kDa in Western blot (WB) .
Applications: Validated for WB, immunofluorescence (IF), immunohistochemistry (IHC), and ELISA .
| Application | Dilution Range |
|---|---|
| Western Blot | 1:1000 – 1:2000 |
| Immunofluorescence | 1:400 – 1:1600 |
| IHC | 1:50 – 1:200 |
Note: Optimal dilution varies by sample type and experimental conditions .
TBC1D25 knockout (KO) mice exhibited exacerbated cardiac hypertrophy and fibrosis after transverse aortic constriction (TAC), with increased expression of hypertrophy markers (ANP, BNP) and fibrosis markers (Collagen Iα, CTGF) . Overexpression of TBC1D25 in cardiomyocytes suppressed angiotensin II (Ang II)-induced hypertrophy by inhibiting TAK1-JNK/p38 signaling .
| Parameter | WT Mice (TAC) | TBC1D25-KO Mice (TAC) |
|---|---|---|
| Cardiomyocyte Size | Moderate | Significantly Larger |
| Fibrosis Area | 15% | 35% |
| ANP/BNP mRNA Levels | Baseline | 3–4x Increase |
TBC1D25 directly interacts with TAK1 via its C-terminal domain (aa 138–226), suppressing TAK1-mediated activation of JNK/p38 pathways. Co-immunoprecipitation (Co-IP) and GST pull-down assays confirmed this interaction .
TBC1D25 (TBC1 domain family, member 25) is a protein containing a TBC domain that functions as a Rab GTPase activating protein (Rab-GAP). It was previously known as ornithine aminotransferase-like 1 (OATL1), though it has no actual similarity to ornithine aminotransferase . The protein plays a critical role in the fusion of autophagosomes with endosomes and lysosomes, making it an essential component of the autophagy pathway . Recent research has shown that TBC1D25 is also upregulated during pathological cardiac remodeling and appears to suppress cardiac hypertrophy, fibrosis, and dysfunction by regulating the TAK1-JNK/p38 signaling pathway . The protein has an observed molecular weight of approximately 80 kDa and is widely expressed in multiple cell types and tissues across human, mouse, and rat species .
Based on the search results, there are at least two main types of TBC1D25 antibodies commercially available:
Monoclonal Antibody (Mouse IgG1):
Polyclonal Antibody:
These antibodies are purified using different methods (Protein G purification for the monoclonal antibody and antigen affinity purification for polyclonal antibodies) and show high sequence identity across species, with 96% identity to mouse and 95% to rat orthologs .
TBC1D25 antibodies show consistent reactivity across multiple mammalian species. The monoclonal antibody (67574-1-Ig) has been tested and confirmed to react with human, mouse, and rat samples . This cross-reactivity is likely due to the high sequence conservation of TBC1D25 across these species. According to the polyclonal antibody information, the immunogen sequence used shows 96% identity to mouse TBC1D25 and 95% identity to rat TBC1D25 .
For Western Blot (WB) applications using TBC1D25 antibody, the following optimized conditions are recommended:
Dilution Range:
Cell Lines Successfully Tested:
The antibody has been validated in multiple cell lines, including:
Protocol Recommendations:
Sample preparation: Standard lysis buffers containing protease inhibitors
Protein loading: 20-30 μg total protein per lane
Blocking: 5% non-fat milk in TBST is typically effective
Incubation: Primary antibody incubation overnight at 4°C gives optimal results
Detection: HRP-conjugated secondary antibody followed by ECL detection
For specific optimization needs, the manufacturer provides specialized WB protocols for this antibody that can be downloaded from their website . It's recommended to perform a dilution series during initial optimization to determine the optimal concentration for your specific experimental conditions.
For Immunofluorescence (IF) applications with TBC1D25 antibody, the following methodology is recommended:
Dilution Range:
Validated Cell Lines:
The antibody has been positively tested for IF in:
Protocol Recommendations:
Cell fixation: 4% paraformaldehyde (15 minutes at room temperature)
Permeabilization: 0.1-0.5% Triton X-100 in PBS (10 minutes)
Blocking: 1-5% BSA or normal serum from the secondary antibody species
Primary antibody incubation: Overnight at 4°C or 1-2 hours at room temperature
Secondary antibody: Fluorophore-conjugated secondary antibody matching the host species (mouse for 67574-1-Ig)
Counterstaining: DAPI for nuclei visualization
Mounting: Anti-fade mounting medium
Essential Controls:
Negative control: Omit primary antibody but include all other steps
Isotype control: Use non-specific mouse IgG1 at the same concentration
Blocking peptide control: Pre-incubate antibody with blocking peptide
Knockdown validation: Use cells with TBC1D25 knockdown or knockout
Subcellular marker co-staining: Co-stain with markers for autophagosomal or endosomal compartments to confirm expected localization
The manufacturer provides specific IF protocols for TBC1D25 antibody that can be downloaded for detailed instructions . Given TBC1D25's role in autophagosome-endosome-lysosome fusion, co-localization studies with markers for these organelles (such as LC3 for autophagosomes or LAMP1 for lysosomes) can provide valuable functional information.
While the search results don't specifically mention immunoprecipitation (IP) protocols for TBC1D25 antibody, we can draw on general principles and information from related TBC family antibodies:
Recommended Considerations:
Antibody Selection:
Monoclonal antibodies like 67574-1-Ig may be suitable for IP due to their high specificity
The antibody should be validated for IP applications before use
Protocol Suggestions:
Lysate preparation: Use mild lysis buffers (e.g., RIPA buffer with protease inhibitors)
Pre-clearing: Pre-clear lysate with Protein G beads to reduce non-specific binding
Antibody binding: Incubate 2-5 μg of antibody with 500-1000 μg of total protein lysate
Capture: Use Protein G magnetic beads for mouse monoclonal antibodies
Incubation time: Overnight at 4°C with gentle rotation
Washing: Multiple washes with decreasing salt concentrations
Elution: Use gentle elution conditions to preserve protein-protein interactions
Controls:
IgG control: Use non-specific mouse IgG1 as a negative control
Input control: Include an aliquot of pre-IP lysate
Known interacting partners: Validate by blotting for documented TBC1D25 interactors
Co-Immunoprecipitation Considerations:
Based on research showing TBC1D25 interacts with TAK1, co-IP experiments should be designed to preserve this interaction
From search result , TBC1D25 directly interacts with TAK1, requiring amino acids 138-226 in the C-terminal region of TBC1D25 and amino acids 1-300 in the C-terminal region of TAK1
Validation Methods:
Western blot after IP to confirm successful precipitation of TBC1D25
Mass spectrometry to identify novel interaction partners
While specific IP protocols for TBC1D25 aren't provided in the search results, these general guidelines combined with the knowledge of TBC1D25's interactions can help design effective IP experiments.
TBC1D25 plays a crucial role in the autophagy pathway as a Rab GTPase-activating protein (Rab-GAP) that specifically regulates the fusion of autophagosomes with endosomes and lysosomes. This function is critical for the completion of autophagy, as it allows for the degradation of autophagosomal contents .
Molecular Mechanism:
TBC1D25 contains a TBC domain, which is characteristic of Rab-GAPs that inactivate Rab GTPases by stimulating their intrinsic GTPase activity, converting them from the active GTP-bound form to the inactive GDP-bound form. This regulation of Rab GTPases is likely how TBC1D25 controls vesicle fusion events in the autophagy pathway.
Experimental Models to Demonstrate Function:
Cellular Models:
Knockout/Knockdown Systems: TALEN-mediated knockout cells can be generated as described for related TBC family members in search result . This approach allows for clean genetic deletion and functional studies.
Overexpression Systems: YFP-tagged or HA-tagged TBC1D25 constructs can be used to study localization and function.
Point Mutations: Creating catalytically inactive mutants (similar to the R381A mutation described for TBC1D17) can help determine if the GAP activity is essential for function .
Autophagy Assays:
LC3 Puncta Formation: Monitor autophagosome formation using fluorescently-tagged LC3.
Autophagic Flux Assays: Use lysosomal inhibitors (e.g., bafilomycin A1) to assess if TBC1D25 affects autophagosome-lysosome fusion.
Long-lived Protein Degradation: Measure the degradation of long-lived proteins as a functional readout of autophagy.
Tandem Fluorescent-tagged LC3 (tfLC3): Use mRFP-GFP-LC3 to monitor autophagosome maturation (GFP fluorescence is quenched in acidic lysosomes while RFP remains stable).
Interaction Studies:
Co-localization Analysis: Immunofluorescence studies can demonstrate co-localization with autophagosomal (LC3), endosomal (Rab5, Rab7), and lysosomal (LAMP1, LAMP2) markers.
Co-immunoprecipitation: Identify Rab GTPases that interact with TBC1D25.
GAP Activity Assays: Measure the GTPase activity of candidate Rab proteins in the presence and absence of TBC1D25.
In Vivo Models:
These experimental approaches can collectively provide a comprehensive understanding of TBC1D25's function in the autophagy pathway and its potential involvement in related pathological conditions.
TBC1D25 plays a significant role in cardiac remodeling as revealed by recent research. According to search result , TBC1D25 is upregulated during pathological cardiac remodeling and functions as a protective factor against adverse cardiac remodeling.
Key Findings and Mechanisms:
Protective Function: TBC1D25 knockout exacerbates cardiac hypertrophy, fibrosis, and dysfunction following partial transverse aortic constriction (TAC), suggesting that TBC1D25 has a cardioprotective role .
Signaling Pathway Regulation: TBC1D25 suppresses pathological cardiac remodeling by regulating the TAK1-JNK/p38 signaling pathway. Specifically:
Direct Interaction: TBC1D25 directly interacts with TAK1, as demonstrated by immunoprecipitation and GST pull-down assays. This interaction requires:
Experimental Approaches for Studying This Pathway:
In Vivo Models:
TAC Model: Partial transverse aortic constriction in TBC1D25-KO mice and wild-type controls to induce cardiac remodeling
Angiotensin II Infusion: An alternative model to induce cardiac hypertrophy and fibrosis
Echocardiography: To assess cardiac function in vivo
Histological Analysis: For assessment of cardiomyocyte size and cardiac fibrosis
In Vitro Models:
H9C2 Cells and NRCMs (Neonatal Rat Cardiomyocytes): Can be used with TBC1D25 overexpression or knockdown
Angiotensin II Treatment: To induce cardiomyocyte hypertrophy in vitro
Cell Size Measurement: As a marker of hypertrophy
Hypertrophic Gene Expression: Analyze expression of markers like ANP, BNP, and β-MHC
Molecular Interaction Studies:
Co-Immunoprecipitation: To confirm TBC1D25-TAK1 interaction
GST Pull-down Assays: To map interaction domains
Mutational Analysis: Creating truncated or point mutants to identify critical residues for interaction
Proximity Ligation Assay (PLA): To visualize protein-protein interactions in situ
Signaling Pathway Analysis:
Western Blotting: To assess phosphorylation levels of TAK1, JNK, and p38 MAPK
Kinase Inhibitors: Use of specific inhibitors (e.g., TAK1 inhibitor 5Z-7-oxozeaenol) to validate pathway involvement
Transcriptional Reporters: To measure downstream effects on transcription factors like AP-1
RNA-seq/Proteomics: To identify global changes in gene/protein expression
Translational Relevance:
Human Heart Samples: Analysis of TBC1D25 expression in heart failure patients
Genetic Association Studies: Examining TBC1D25 polymorphisms in relation to heart disease
Therapeutic Targeting: Testing compounds that modulate the TBC1D25-TAK1 interaction
This multi-faceted approach allows researchers to comprehensively study the role of TBC1D25 in cardiac remodeling, from molecular mechanisms to potential therapeutic applications.
TBC1D25 antibodies can serve as valuable tools for investigating autophagosome-lysosome fusion events, given the protein's role in this critical step of the autophagy pathway. Here are methodological approaches for using these antibodies to study fusion dynamics:
1. Co-localization Studies:
Confocal Microscopy Approach:
Use TBC1D25 antibody (67574-1-Ig at 1:400-1:600 dilution) alongside markers for autophagosomes (LC3), late endosomes (Rab7), and lysosomes (LAMP1/2)
Quantify co-localization coefficients (Pearson's or Mander's) between TBC1D25 and these markers under various conditions
Time-lapse imaging can capture dynamic fusion events with fluorescently-tagged proteins
Super-resolution Microscopy:
Techniques like STORM or STED can provide nanoscale resolution of TBC1D25 localization relative to fusion machinery
Particularly useful for resolving the precise spatial arrangement of TBC1D25 at fusion sites
2. Functional Assays:
Tandem Fluorescent-tagged Reporters:
Use mRFP-GFP-LC3 to monitor autophagosome maturation while simultaneously immunostaining for TBC1D25
In unfused autophagosomes, both GFP and RFP signals are visible; after fusion with lysosomes, the acidic environment quenches GFP but not RFP
Correlation between TBC1D25 presence and fusion events can be quantified
Lysosomal Enzyme Delivery Assays:
Monitor the delivery of lysosomal enzymes to autophagosomes in relation to TBC1D25 expression levels
Cathepsin activity within autophagosomes can be measured using activity-based probes
3. Molecular Interaction Analysis:
Proximity Ligation Assay (PLA):
Detect in situ interactions between TBC1D25 and components of the fusion machinery
This technique can visualize protein interactions within 40 nm distance in fixed cells
FRET/BRET Analysis:
For live cell studies, combine antibody validation with fluorescent protein-tagged constructs
Measure energy transfer between TBC1D25 and fusion machinery components
4. Manipulation Strategies:
Knockdown/Knockout with Rescue Experiments:
Domain-specific Mutations:
Create mutations in TBC1D25's Rab-GAP domain to assess functional importance
Use antibodies to track localization of these mutants relative to fusion sites
5. Biochemical Fractionation:
Isolation of Autophagosomes:
6. Disease-relevant Models:
Cardiac Hypertrophy Models:
These methodological approaches provide a comprehensive framework for using TBC1D25 antibodies to study autophagosome-lysosome fusion events in normal physiology and disease states.
When working with TBC1D25 antibody in Western blot applications, researchers may encounter several common issues. Here are problems that might arise and their recommended solutions:
1. Weak or No Signal:
2. High Background:
3. Multiple Bands or Unexpected Band Size:
| Problem | Possible Causes | Solutions |
|---|---|---|
| Protein degradation | Sample degradation | Add fresh protease inhibitors, keep samples cold |
| Post-translational modifications | Different protein states | Use phosphatase inhibitors if phosphorylation is suspected |
| Splice variants | Alternative splicing of TBC1D25 | Verify with literature if splice variants exist |
| Non-specific binding | Insufficient blocking | Optimize blocking conditions |
| Sample overloading | Too much protein | Reduce total protein load |
4. Optimization Strategies:
5. Sample-specific Considerations:
Based on validation data, the TBC1D25 antibody (67574-1-Ig) works well in multiple cell lines including NIH/3T3, HeLa, HEK-293, Jurkat, HSC-T6, and Neuro-2a cells . If working with other cell types or tissues, additional optimization may be necessary. Consider using one of these validated cell lines as a positive control during troubleshooting.
For detailed, product-specific protocols, the manufacturer provides a downloadable WB protocol specifically for TBC1D25 antibody 67574-1-Ig that may contain additional optimization tips .
Validating antibody specificity is crucial for ensuring reliable and reproducible research findings. For TBC1D25 antibody, several validation strategies can be employed:
1. Genetic Validation Approaches:
2. Peptide Competition Assay:
Pre-incubate the antibody with excess immunizing peptide or recombinant TBC1D25 protein
Apply the neutralized antibody in parallel with regular antibody
Expected outcome: Significant reduction or elimination of specific signal with neutralized antibody
3. Cross-validation with Multiple Antibodies:
4. Mass Spectrometry Validation:
Perform immunoprecipitation with TBC1D25 antibody
Analyze precipitated proteins by mass spectrometry
Confirm presence of TBC1D25 peptides in precipitated material
5. Orthogonal Methods for Functional Validation:
6. Tissue/Cell Type-Specific Controls:
Include both positive and negative control samples in experiments
Positive controls: Cell lines with confirmed TBC1D25 expression (NIH/3T3, HeLa, HEK-293, Jurkat, HSC-T6, Neuro-2a)
Negative controls: TBC1D25 knockout cells or tissues with minimal expression
7. Recombinant Protein Standards:
Use purified recombinant TBC1D25 protein at known concentrations
Create standard curve for quantitative applications
Verify antibody detection limit and linear range
8. Isotype Control:
For immunostaining experiments, include isotype control (mouse IgG1 for 67574-1-Ig)
Process identically to experimental samples
Helps distinguish specific from non-specific binding
These validation approaches, especially when used in combination, provide robust evidence for antibody specificity and support confidence in experimental results obtained with TBC1D25 antibodies.
Sample preparation has significant effects on antibody performance across different applications. For TBC1D25 antibody, optimizing sample preparation is crucial for obtaining reliable and consistent results:
1. Western Blot Sample Preparation:
2. Immunofluorescence/ICC Sample Preparation:
3. Immunohistochemistry Sample Preparation:
4. Immunoprecipitation Sample Preparation:
| Parameter | Recommendations | Rationale |
|---|---|---|
| Lysis buffer | Non-denaturing buffer (e.g., NP-40 buffer) | Preserves protein-protein interactions |
| Cell/tissue amount | 1-3 mg total protein | Sufficient to detect interactions |
| Pre-clearing | Incubate lysate with beads prior to antibody | Reduces non-specific binding |
| Antibody amount | 2-5 μg per mg of protein | Ensure sufficient antibody for target capture |
| Controls | Include IgG control and input samples | Essential for specificity assessment |
5. Critical Factors Affecting Performance Across Applications:
| Factor | Effect | Recommendation |
|---|---|---|
| Protein degradation | Loss of epitopes | Use fresh samples with protease inhibitors |
| Post-translational modifications | May mask or expose epitopes | Consider phosphatase inhibitors if studying signaling |
| Fixation cross-linking | Can mask epitopes | Optimize fixation time and antigen retrieval |
| Sample storage | Freeze-thaw can degrade proteins | Aliquot samples to avoid repeated freeze-thaw cycles |
| Antibody batch variation | Different lots may have varying sensitivity | Include standard positive controls with each experiment |
6. Application-Specific Considerations for TBC1D25:
For studying TBC1D25's role in autophagy, sample preparation should account for autophagy dynamics:
Autophagy induction (starvation, rapamycin) or inhibition (bafilomycin A1) protocols
Time-course experiments to capture dynamic processes
Co-immunoprecipitation buffers that preserve interactions with TAK1 and other signaling components
Optimizing sample preparation based on these recommendations will significantly improve the reliability and reproducibility of experiments using TBC1D25 antibodies across different applications.
TBC1D25 antibodies can be valuable tools for investigating the protein's involvement in various disease models, particularly given its roles in autophagy and cardiac remodeling. Here are methodological approaches for different disease contexts:
1. Cardiovascular Diseases:
Based on search result , TBC1D25 plays a protective role against pathological cardiac remodeling, providing a strong foundation for cardiovascular research applications.
2. Neurodegenerative Disorders:
Given TBC1D25's role in autophagy and the importance of autophagy defects in neurodegenerative diseases:
| Disease Model | Experimental Approach | Antibody Application |
|---|---|---|
| Alzheimer's disease | Human brain tissue samples or transgenic mouse models | IHC to correlate TBC1D25 expression with amyloid pathology |
| Parkinson's disease | MPTP-treated or α-synuclein transgenic mice | WB and IF to assess alterations in dopaminergic neurons |
| Autophagy impairment models | Bafilomycin A1 or chloroquine treatment | IF to monitor autophagosome-lysosome fusion defects |
| mTOR inhibition | Rapamycin treatment | WB to assess TBC1D25 expression during autophagy induction |
3. Cancer Research:
Autophagy has context-dependent roles in cancer, making TBC1D25 a potential target for investigation:
4. Inflammation and Immunity:
TBC1D25's connection to TAK1 signaling suggests potential roles in inflammation:
5. Methodological Strategies Across Disease Models:
The TBC1D25 antibodies described in the search results (particularly 67574-1-Ig and PA5-56082) have been validated in multiple cell types and applications , making them suitable for these diverse disease research contexts. By applying these antibodies within the suggested experimental frameworks, researchers can gain valuable insights into TBC1D25's contributions to disease pathogenesis and potential therapeutic interventions.
Based on the search results and emerging research directions, several key trends and methodological innovations are shaping TBC1D25 research:
1. Signaling Pathway Integration:
One of the most significant recent findings is TBC1D25's involvement in the TAK1-JNK/p38 MAPK signaling pathway, particularly in cardiac remodeling . This opens several new research directions:
| Research Trend | Methodological Approach | Significance |
|---|---|---|
| Multi-omics integration | Combine phosphoproteomics with TBC1D25 antibody-based studies | Maps the complete signaling network |
| Proximity-based proteomics | BioID or APEX2 tagging of TBC1D25 | Identifies novel interaction partners |
| Single-cell signaling analysis | Antibody-based imaging with phospho-specific markers | Reveals cell-to-cell variability in response |
| Computational modeling | Integration of TBC1D25-TAK1-JNK/p38 pathway kinetics | Predicts intervention points in disease |
2. Autophagy Regulation Mechanisms:
TBC1D25's role in autophagosome-endosome-lysosome fusion continues to be an active area of research:
| Research Trend | Methodological Approach | Significance |
|---|---|---|
| Live-cell autophagy dynamics | CRISPR-based endogenous tagging with fluorescent proteins | Real-time visualization of native TBC1D25 |
| Super-resolution microscopy | dSTORM or STED imaging with TBC1D25 antibodies | Nanoscale localization during fusion events |
| Correlation with Rab activation status | FRET-based Rab activity sensors | Determines how TBC1D25 affects specific Rab GTPases |
| Selective autophagy pathways | Co-localization with cargo-specific receptors | Reveals pathway-specific roles |
3. Cross-talk Between Autophagy and Inflammation:
The connection between TBC1D25, TAK1 signaling, and autophagy suggests important cross-talk:
| Research Trend | Methodological Approach | Significance |
|---|---|---|
| Inflammasome regulation | TBC1D25 antibody studies in NLRP3 activation models | Links autophagy to inflammatory responses |
| Mitophagy-inflammation axis | Dual labeling of mitochondria and TBC1D25 | Reveals role in mitochondrial quality control |
| NF-κB pathway interaction | ChIP-seq after TBC1D25 modulation | Identifies transcriptional consequences |
| Cytokine production profiling | Multiplex assays with TBC1D25 manipulation | Quantifies impact on inflammatory output |
4. Advanced Genetic Tools:
The development of precise genetic tools enhances TBC1D25 functional studies:
| Research Trend | Methodological Approach | Significance |
|---|---|---|
| Domain-specific CRISPR editing | Precise modification of TBC domain | Structure-function analysis without complete knockout |
| Conditional knockout models | Tissue-specific TBC1D25 deletion | Addresses function in specific physiological contexts |
| Base editing | Introduction of specific mutations | Creates models of potential human variants |
| Inducible expression systems | Temporal control of TBC1D25 expression | Studies acute vs. chronic effects |
5. Translational Research Applications:
Moving TBC1D25 research toward clinical applications:
6. Emerging Technology Integration:
| Research Trend | Methodological Approach | Significance |
|---|---|---|
| Organ-on-chip technology | TBC1D25 monitoring in 3D cardiac models | More physiologically relevant than 2D culture |
| AI-based image analysis | Deep learning for TBC1D25 localization patterns | Identifies subtle phenotypes in large datasets |
| Spatial transcriptomics | Correlation of TBC1D25 protein with local gene expression | Reveals microenvironmental influences |
| Cryo-electron tomography | Structural visualization of TBC1D25 in native context | Molecular mechanism insights |
These emerging trends and methodological approaches represent the cutting edge of TBC1D25 research, driven by its newly discovered roles in signaling and disease processes. The antibodies described in the search results (67574-1-Ig, PA5-56082) will be essential tools for many of these applications , particularly when integrated with these advanced methodological approaches.