TBC1D25 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of TBC1D25 Antibody

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 .

Key Features of TBC1D25 Antibody

  • 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 .

Recommended Dilutions

ApplicationDilution Range
Western Blot1:1000 – 1:2000
Immunofluorescence1:400 – 1:1600
IHC1:50 – 1:200

Note: Optimal dilution varies by sample type and experimental conditions .

Validated Cell Lines

  • WB: NIH/3T3, HeLa, HEK-293, Jurkat, HSC-T6, Neuro-2a .

  • IF/ICC: NIH/3T3, HEK-293 .

Role in Cardiac Remodeling

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 .

Key Data from International Journal of Biological Sciences (2020):

ParameterWT Mice (TAC)TBC1D25-KO Mice (TAC)
Cardiomyocyte SizeModerateSignificantly Larger
Fibrosis Area15%35%
ANP/BNP mRNA LevelsBaseline3–4x Increase

Mechanism of Action

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 .

Limitations and Cautions

  • Research Use Only: Not validated for diagnostic or therapeutic purposes .

  • Batch Variability: Performance may differ between lots; recommend validation for novel applications .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
MG81 antibody; MGC126866 antibody; MGC126868 antibody; MGC149731 antibody; MGC149732 antibody; OATL1 antibody; ORNITHINE AMINOTRANSFERASE LIKE 1 antibody; TBC1 domain family member 25 antibody; TBC1D25 antibody; TBC25_HUMAN antibody
Target Names
TBC1D25
Uniprot No.

Target Background

Function
TBC1D25 Antibody acts as a GTPase-activating protein specific for RAB33B. It plays a role in regulating autophagosome maturation, the process where autophagosomes fuse with endosomes and lysosomes.
Gene References Into Functions
  1. Our study demonstrated that adenovirus-mediated antisense ODC expression inhibits tumor cell growth by blocking the polyamine synthesis pathway. PMID: 17558447
  2. Research indicates that each of the four genes investigated was associated with at least one major outcome: anxiety (SAT1, SMS), mood disorders (SAT1, SMOX), and suicide attempts (SAT1, OATL1). PMID: 21152090
Database Links

HGNC: 8092

OMIM: 311240

KEGG: hsa:4943

STRING: 9606.ENSP00000365962

UniGene: Hs.694353

Subcellular Location
Cytoplasm. Cytoplasmic vesicle, autophagosome. Note=It is dispersed in the cytoplasm under nutrient-rich conditions. Localizes at autophagosomes under cell starving conditions.

Q&A

What is TBC1D25 and what cellular functions does it regulate?

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 .

What are the available types of TBC1D25 antibodies and their source organisms?

Based on the search results, there are at least two main types of TBC1D25 antibodies commercially available:

  • Monoclonal Antibody (Mouse IgG1):

    • Clone designation: 2C7H3

    • Product number: 67574-1-Ig

    • Host organism: Mouse

    • Format: Unconjugated, liquid form

    • Storage buffer: PBS with 0.02% sodium azide and 50% glycerol (pH 7.3)

  • Polyclonal Antibody:

    • Product number: PA5-56082

    • Host organism: Rabbit

    • Format: Unconjugated

    • Immunogen sequence: AVDPQITSLD VLQHILIRAF DLSGKKNFGI SYLGRDRLGQ EVYLSLLSDW DLSTAFATAS KPYLQLRVDI RPSEDSPLLE DWDIISPKDV SGSDVLLAEK RSSLTTAALP FTQSIL

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 .

What is the reactivity profile of TBC1D25 antibodies across species?

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 .

What are the optimal conditions for using TBC1D25 antibody in Western Blot applications?

For Western Blot (WB) applications using TBC1D25 antibody, the following optimized conditions are recommended:

Dilution Range:

  • For monoclonal antibody (67574-1-Ig): 1:1000-1:2000 dilution

Cell Lines Successfully Tested:
The antibody has been validated in multiple cell lines, including:

  • NIH/3T3 cells

  • HeLa cells

  • HEK-293 cells

  • Jurkat cells

  • HSC-T6 cells

  • Neuro-2a cells

Protocol Recommendations:

  • Sample preparation: Standard lysis buffers containing protease inhibitors

  • Protein loading: 20-30 μg total protein per lane

  • Expected band size: 80 kDa (observed molecular weight)

  • 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.

How should TBC1D25 antibody be used for Immunofluorescence and what controls are essential?

For Immunofluorescence (IF) applications with TBC1D25 antibody, the following methodology is recommended:

Dilution Range:

  • For monoclonal antibody (67574-1-Ig): 1:400-1:1600 dilution

Validated Cell Lines:
The antibody has been positively tested for IF in:

  • NIH/3T3 cells

  • HEK-293 cells

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.

What considerations should be made when using TBC1D25 antibody for immunoprecipitation studies?

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.

How does TBC1D25 function in the autophagy pathway and what experimental models best demonstrate this?

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:

    • TBC1D25-KO Mice: As mentioned in search result , TBC1D25-KO mice have been generated and used to study cardiac remodeling. These mice could also be useful for studying the role of TBC1D25 in autophagy in different tissues and under various stress conditions.

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.

What is the role of TBC1D25 in cardiac remodeling and how can researchers study this pathway?

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:

    • TBC1D25 deficiency increases phosphorylation levels of TAK1, JNK, and p38

    • Overexpression of TBC1D25 inhibits phosphorylation of these proteins

    • TAK1 appears to be the key molecule in this regulatory process

  • Direct Interaction: TBC1D25 directly interacts with TAK1, as demonstrated by immunoprecipitation and GST pull-down assays. This interaction requires:

    • Amino acids 138-226 in the C-terminal region of TBC1D25

    • Amino acids 1-300 in the C-terminal region of TAK1

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.

How can TBC1D25 antibodies be used to study autophagosome-lysosome fusion events?

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:

    • Generate TBC1D25-deficient cells using TALEN technology as described in result

    • Rescue with wild-type or mutant TBC1D25 constructs

    • Use antibodies to confirm expression levels in 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:

    • Perform subcellular fractionation to isolate autophagosomal, endosomal, and lysosomal fractions

    • Use Western blotting with TBC1D25 antibody (67574-1-Ig at 1:1000-1:2000) to determine distribution across fractions

    • Analyze how distribution changes during induction of autophagy or inhibition of fusion

6. Disease-relevant Models:

  • Cardiac Hypertrophy Models:

    • Leverage the cardiac protection role of TBC1D25 (from result ) to study how autophagy fusion events may be impacted during cardiac stress

    • Use the TBC1D25 antibody to assess expression changes in response to stress conditions

These methodological approaches provide a comprehensive framework for using TBC1D25 antibodies to study autophagosome-lysosome fusion events in normal physiology and disease states.

What are common issues with TBC1D25 antibody in Western blot and how can they be resolved?

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:

ProblemPossible CausesSolutions
Insufficient proteinLow expression of TBC1D25 in sampleIncrease protein loading (30-50 μg)
Insufficient antibodyDilution too highTry more concentrated antibody (1:500-1:1000)
Inadequate transferInefficient protein transferOptimize transfer conditions for high MW proteins (80 kDa)
Degraded antibodyImproper storageAliquot antibody and store at -20°C; avoid repeated freeze-thaw cycles
Blocking interferenceBlocking agent masks epitopeTry different blocking agents (BSA vs. milk)

2. High Background:

ProblemPossible CausesSolutions
Non-specific bindingAntibody concentration too highIncrease dilution (1:2000-1:5000)
Inadequate blockingInsufficient blockingIncrease blocking time (2 hours) or concentration (5%)
Insufficient washingResidual unbound antibodyIncrease number and duration of wash steps
Cross-reactivityAntibody recognizes related proteinsUse alternative TBC1D25 antibody (e.g., polyclonal PA5-56082)
Buffer contaminationBacterial growth in buffersPrepare fresh buffers with sterile water

3. Multiple Bands or Unexpected Band Size:

ProblemPossible CausesSolutions
Protein degradationSample degradationAdd fresh protease inhibitors, keep samples cold
Post-translational modificationsDifferent protein statesUse phosphatase inhibitors if phosphorylation is suspected
Splice variantsAlternative splicing of TBC1D25Verify with literature if splice variants exist
Non-specific bindingInsufficient blockingOptimize blocking conditions
Sample overloadingToo much proteinReduce total protein load

4. Optimization Strategies:

ParameterRange to TestNotes
Antibody dilution1:500 to 1:5000Start with recommended 1:1000-1:2000
Blocking agent3-5% milk or BSATry both to determine optimal
Blocking time1-3 hours or overnightLonger may reduce background
Primary antibody incubation1-3 hours RT or overnight at 4°COvernight at 4°C often gives best results
Secondary antibody dilution1:2000 to 1:10000Higher dilutions can reduce background
Washing bufferTBST (0.05-0.1% Tween)Multiple washes of 5-10 minutes each

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 .

How can researchers validate the specificity of TBC1D25 antibody results?

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:

ApproachMethodologyExpected Outcome
CRISPR/Cas9 knockoutGenerate TBC1D25 knockout cell lines using CRISPR/Cas9 or TALEN technology Absence of band in knockout cells by Western blot or immunostaining
siRNA/shRNA knockdownTransfect cells with TBC1D25-specific siRNA or shRNASignificant reduction in signal intensity
Rescue experimentsReintroduce TBC1D25 in knockout cellsRestoration of signal at expected molecular weight
OverexpressionTransfect cells with TBC1D25 expression plasmidIncreased signal intensity at expected molecular weight

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:

ApproachMethodologyExpected Outcome
Multiple antibody comparisonCompare monoclonal (67574-1-Ig) with polyclonal (PA5-56082) TBC1D25 antibodies Consistent detection pattern with both antibodies
Different epitope antibodiesUse antibodies recognizing different epitopes of TBC1D25Concordant results indicate higher specificity
Different detection methodsCompare results from WB, IHC, and IF applicationsConsistent localization and expression patterns

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:

ApproachMethodologyExpected Outcome
qRT-PCR correlationCompare protein levels (antibody) with mRNA levelsConcordant expression patterns
Known interactor co-localizationCo-stain for TBC1D25 and TAK1 Expected co-localization pattern
Functional assaysAssess impact on autophagy or TAK1 signaling Functional outcome should align with expression data

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.

How does sample preparation affect TBC1D25 antibody performance in different applications?

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:

ParameterRecommendationsRationale
Lysis bufferRIPA buffer with protease inhibitorsEfficiently extracts TBC1D25 while maintaining protein integrity
Lysis conditionsKeep samples cold (4°C)Prevents protein degradation
Protein denaturationHeat at 95°C for 5 min in Laemmli bufferComplete denaturation improves epitope accessibility
Protein amount20-30 μg for cell lines with known expression Sufficient to detect the 80 kDa TBC1D25 protein
Gel percentage8-10% SDS-PAGEOptimal separation for 80 kDa proteins
Transfer conditionsWet transfer, 100V for 60-90 minutesComplete transfer of high molecular weight proteins

2. Immunofluorescence/ICC Sample Preparation:

ParameterRecommendationsRationale
Fixation4% paraformaldehyde, 10-15 minutesPreserves protein structure while allowing antibody access
Alternative fixationMethanol (-20°C, 10 min) for epitope retrievalMay expose epitopes masked by PFA fixation
Permeabilization0.1-0.5% Triton X-100, 5-10 minutesAllows antibody access to intracellular targets
Blocking5% normal serum matching secondary antibody speciesReduces non-specific binding
Cell density50-70% confluenceAllows clear visualization of subcellular structures
Validated cell linesNIH/3T3, HEK-293 Confirmed to express detectable TBC1D25 levels

3. Immunohistochemistry Sample Preparation:

ParameterRecommendationsRationale
Fixation10% neutral buffered formalinStandard fixative for tissue preservation
Antigen retrievalTE buffer pH 9.0 (heat-induced epitope retrieval)Recommended for optimal epitope exposure
Alternative retrievalCitrate buffer pH 6.0 as alternativeMay be effective if TE buffer gives insufficient results
Section thickness4-5 μmThin enough for good reagent penetration
BlockingHydrogen peroxide followed by serum blockBlocks endogenous peroxidase and prevents non-specific binding
Tissue typesCardiac tissue for TBC1D25 function studies Relevant for cardiac remodeling research

4. Immunoprecipitation Sample Preparation:

ParameterRecommendationsRationale
Lysis bufferNon-denaturing buffer (e.g., NP-40 buffer)Preserves protein-protein interactions
Cell/tissue amount1-3 mg total proteinSufficient to detect interactions
Pre-clearingIncubate lysate with beads prior to antibodyReduces non-specific binding
Antibody amount2-5 μg per mg of proteinEnsure sufficient antibody for target capture
ControlsInclude IgG control and input samplesEssential for specificity assessment

5. Critical Factors Affecting Performance Across Applications:

FactorEffectRecommendation
Protein degradationLoss of epitopesUse fresh samples with protease inhibitors
Post-translational modificationsMay mask or expose epitopesConsider phosphatase inhibitors if studying signaling
Fixation cross-linkingCan mask epitopesOptimize fixation time and antigen retrieval
Sample storageFreeze-thaw can degrade proteinsAliquot samples to avoid repeated freeze-thaw cycles
Antibody batch variationDifferent lots may have varying sensitivityInclude 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.

How can TBC1D25 antibodies be used to investigate its role in disease models?

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.

Disease ModelExperimental ApproachAntibody Application
Pressure overload-induced cardiac hypertrophyTransverse aortic constriction (TAC) in animal models WB (1:1000) to monitor TBC1D25 expression changes during disease progression
Angiotensin II-induced cardiomyocyte hypertrophyIn vitro treatment of H9C2 cells or NRCMs IF (1:400-1:600) to visualize subcellular localization changes
Ischemia-reperfusion injuryEx vivo Langendorff heart preparationIHC to assess regional expression changes
Heart failureHuman heart failure tissue samplesIHC and WB to compare TBC1D25 levels between normal and failing hearts

2. Neurodegenerative Disorders:

Given TBC1D25's role in autophagy and the importance of autophagy defects in neurodegenerative diseases:

Disease ModelExperimental ApproachAntibody Application
Alzheimer's diseaseHuman brain tissue samples or transgenic mouse modelsIHC to correlate TBC1D25 expression with amyloid pathology
Parkinson's diseaseMPTP-treated or α-synuclein transgenic miceWB and IF to assess alterations in dopaminergic neurons
Autophagy impairment modelsBafilomycin A1 or chloroquine treatmentIF to monitor autophagosome-lysosome fusion defects
mTOR inhibitionRapamycin treatmentWB to assess TBC1D25 expression during autophagy induction

3. Cancer Research:

Autophagy has context-dependent roles in cancer, making TBC1D25 a potential target for investigation:

Cancer ContextExperimental ApproachAntibody Application
Tumor cell linesCompare TBC1D25 expression across cancer cell linesWB (1:1000-1:2000) for expression profiling
Chemotherapy resistancePre/post-treatment modelsIF to assess autophagy flux changes
Nutrient deprivationGlucose or amino acid starvationWB to monitor TBC1D25 response to metabolic stress
Tumor tissue microarraysPatient samples across cancer stagesIHC to correlate expression with prognosis

4. Inflammation and Immunity:

TBC1D25's connection to TAK1 signaling suggests potential roles in inflammation:

ContextExperimental ApproachAntibody Application
NF-κB signalingTNF-α or IL-1β stimulationWB to assess TBC1D25 dynamics during inflammatory signaling
Macrophage polarizationM1/M2 polarization protocolsIF (1:400-1:1600) to visualize localization changes
Autoimmune modelsExperimental autoimmune encephalomyelitisIHC in tissue sections to assess inflammation correlation
Bacterial infectionPathogen challenge in cells/tissuesIF for co-localization with bacterial compartments

5. Methodological Strategies Across Disease Models:

ApproachImplementationAdvantages
Multi-antibody validationUse both monoclonal (67574-1-Ig) and polyclonal (PA5-56082) antibodies Confirms specificity of findings
Genetic manipulationCRISPR/TALEN knockout with antibody validation Establishes causality in disease processes
Rescue experimentsRe-introduce wild-type or mutant TBC1D25Maps functional domains relevant to disease
Systems biologyCombine with transcriptomics/proteomicsPlaces TBC1D25 in broader disease networks
Drug screeningMonitor TBC1D25 expression/localization after compound treatmentIdentifies modulators for therapeutic development

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.

What are the latest research trends involving TBC1D25 and what methodological approaches are being developed?

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 TrendMethodological ApproachSignificance
Multi-omics integrationCombine phosphoproteomics with TBC1D25 antibody-based studiesMaps the complete signaling network
Proximity-based proteomicsBioID or APEX2 tagging of TBC1D25Identifies novel interaction partners
Single-cell signaling analysisAntibody-based imaging with phospho-specific markersReveals cell-to-cell variability in response
Computational modelingIntegration of TBC1D25-TAK1-JNK/p38 pathway kineticsPredicts 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 TrendMethodological ApproachSignificance
Live-cell autophagy dynamicsCRISPR-based endogenous tagging with fluorescent proteinsReal-time visualization of native TBC1D25
Super-resolution microscopydSTORM or STED imaging with TBC1D25 antibodiesNanoscale localization during fusion events
Correlation with Rab activation statusFRET-based Rab activity sensorsDetermines how TBC1D25 affects specific Rab GTPases
Selective autophagy pathwaysCo-localization with cargo-specific receptorsReveals pathway-specific roles

3. Cross-talk Between Autophagy and Inflammation:

The connection between TBC1D25, TAK1 signaling, and autophagy suggests important cross-talk:

Research TrendMethodological ApproachSignificance
Inflammasome regulationTBC1D25 antibody studies in NLRP3 activation modelsLinks autophagy to inflammatory responses
Mitophagy-inflammation axisDual labeling of mitochondria and TBC1D25Reveals role in mitochondrial quality control
NF-κB pathway interactionChIP-seq after TBC1D25 modulationIdentifies transcriptional consequences
Cytokine production profilingMultiplex assays with TBC1D25 manipulationQuantifies impact on inflammatory output

4. Advanced Genetic Tools:

The development of precise genetic tools enhances TBC1D25 functional studies:

Research TrendMethodological ApproachSignificance
Domain-specific CRISPR editingPrecise modification of TBC domainStructure-function analysis without complete knockout
Conditional knockout modelsTissue-specific TBC1D25 deletionAddresses function in specific physiological contexts
Base editingIntroduction of specific mutationsCreates models of potential human variants
Inducible expression systemsTemporal control of TBC1D25 expressionStudies acute vs. chronic effects

5. Translational Research Applications:

Moving TBC1D25 research toward clinical applications:

Research TrendMethodological ApproachSignificance
Biomarker developmentAntibody-based assays for TBC1D25 in patient samplesPotential diagnostic applications
Small molecule screeningHigh-content imaging with TBC1D25 antibodiesIdentifies modulators for therapeutic development
Gene therapy approachesAAV-mediated TBC1D25 deliveryPotential treatment for cardiac conditions
Patient-derived modelsiPSC-derived cardiomyocytes with TBC1D25 modulationPersonalized medicine applications

6. Emerging Technology Integration:

Research TrendMethodological ApproachSignificance
Organ-on-chip technologyTBC1D25 monitoring in 3D cardiac modelsMore physiologically relevant than 2D culture
AI-based image analysisDeep learning for TBC1D25 localization patternsIdentifies subtle phenotypes in large datasets
Spatial transcriptomicsCorrelation of TBC1D25 protein with local gene expressionReveals microenvironmental influences
Cryo-electron tomographyStructural visualization of TBC1D25 in native contextMolecular 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.

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.