SMAC/DIABLO antibodies are immunoassays designed to detect and quantify the Smac/DIABLO protein in experimental settings. These antibodies facilitate studies on its apoptotic function—neutralizing inhibitor of apoptosis proteins (IAPs) to activate caspases—and its non-canonical roles in lipid synthesis and cancer progression .
Commercial SMAC/DIABLO antibodies vary in host species, reactivity, and applications. Below is a comparative table of widely used antibodies:
WB: Western blotting detects Smac/DIABLO at ~21–27 kDa, though observed molecular weights may vary due to post-translational modifications .
IHC/ICC: Antibodies like ab111893 localize Smac/DIABLO in mitochondria and cytoplasm .
Mitochondrial Release: SMAC/DIABLO antibodies confirmed its translocation from mitochondria to cytosol during apoptosis induced by UV, γ-irradiation, or death receptor activation . Bcl-2 overexpression inhibits this release, linking mitochondrial permeability to apoptotic regulation .
Caspase Activation: Antibodies demonstrated that Smac/DIABLO promotes procaspase-3 activation by displacing XIAP from caspase-9 .
Lipid Synthesis: SMAC/DIABLO binds mitochondrial phosphatidylserine decarboxylase (PSD), inhibiting phosphatidylethanolamine synthesis. Depletion via siRNA reduces phospholipid levels and cancer cell proliferation .
Immune Modulation: Proteomics revealed SMAC/DIABLO’s association with neutrophil-mediated immunity and vesicular transport in lung cancer cells .
Ubiquitous Expression: SMAC/DIABLO is detected in liver, kidney, lung, and testes, with high abundance in germinal and parenchymal cells .
Buffer Compatibility: Antibodies like MA1-936 require reduced salt concentrations (0.15 M NaCl) for cross-reactivity in rodent samples .
Validation: Specificity is confirmed via knockout cell lines or competitive assays. For example, ab111893 shows no cross-reactivity in SMAC/DIABLO-depleted A549 cells .
SMAC/DIABLO antibodies underpin efforts to target IAPs in cancer therapy. For example, SMAC mimetics combined with chemotherapeutics enhance apoptosis in resistant tumors .
The antibody is supplied at a concentration of 1mg/ml in a solution of PBS at pH 7.4, supplemented with 10% glycerol and 0.02% sodium azide.
Diablo homolog mitochondrial, Second mitochondria-derived activator of caspase, Smac protein, Direct IAP-binding protein with low pI, DIABLO, SMAC, SMAC3, DIABLO-S, FLJ10537, FLJ25049.
SMAC/DIABLO (Second Mitochondria-derived Activator of Caspases/Direct IAP Binding protein with Low pI) is a mitochondrial protein that plays a crucial role in promoting apoptosis by neutralizing members of the Inhibitor of Apoptosis Protein (IAP) family. The protein is synthesized as a 239 amino acid precursor (~27 kDa) containing a mitochondrial targeting signal peptide in the first 55 amino acids. Upon import into mitochondria, this signal peptide is cleaved to generate the mature Smac/DIABLO protein (~21 kDa). During apoptotic events, SMAC/DIABLO is released from mitochondria into the cytosol where it antagonizes IAP inhibitory effects on both initiator and effector caspases, thereby promoting cell death. This antagonistic activity is based on its N-terminal tetrapeptide (AVPI) that binds to baculoviral IAP repeat (BIR) domains of IAPs.
SMAC/DIABLO antibodies are utilized across multiple experimental techniques in apoptosis research. The primary applications include Western Blot (WB) analysis for protein expression quantification, Immunofluorescence (IF) and Immunocytochemistry (ICC) for subcellular localization studies, Immunohistochemistry (IHC) for tissue expression patterns, Immunoprecipitation (IP) for protein-protein interaction studies, Flow Cytometry (FC) for intracellular detection, and Enzyme-Linked Immunosorbent Assay (ELISA) for protein quantification. These methods allow researchers to investigate SMAC/DIABLO's role in apoptotic pathways, particularly its translocation from mitochondria to cytosol during cell death processes.
For accurately detecting SMAC/DIABLO translocation during apoptosis, subcellular fractionation is the gold standard methodology. Begin with careful mitochondrial isolation using either differential centrifugation or commercial mitochondrial isolation kits. For Western blot analysis, simultaneously prepare cytosolic fractions from control and apoptosis-induced samples. The quality of fractionation should be verified using markers for mitochondria (e.g., COX IV) and cytosol (e.g., GAPDH). When processing samples, maintain cold temperatures (4°C) and include protease inhibitors to prevent degradation. For immunofluorescence approaches, fixation timing is critical – over-fixation may mask epitopes while under-fixation risks losing cellular architecture. Optimal results are typically achieved with 4% paraformaldehyde fixation for 15-20 minutes followed by careful permeabilization with 0.1-0.2% Triton X-100. Co-staining with mitochondrial markers (such as MitoTracker or TOM20) allows precise assessment of SMAC/DIABLO translocation from mitochondria to cytosol during apoptotic events.
Optimizing antibody dilutions is essential for generating reliable, reproducible data with SMAC/DIABLO antibodies across different applications. For Western blot analysis, a titration approach is recommended starting with the manufacturer's suggested range (e.g., 1:5000-1:50000 for monoclonal antibodies or 1:2000-1:16000 for polyclonal antibodies). For immunofluorescence and immunocytochemistry applications, begin with middle-range dilutions (e.g., 1:400-1:500) and adjust based on signal-to-noise ratio. Flow cytometry typically requires higher concentrations – approximately 0.40 μg per 10^6 cells in a 100 μl suspension is a good starting point. Immunoprecipitation protocols should be calibrated using 0.5-4.0 μg of antibody for every 1.0-3.0 mg of total protein lysate. Each new lot of antibody should undergo optimization, even if using previously established protocols, as batch-to-batch variation can significantly impact performance. Additionally, different cell lines or tissue types may require adjusted dilutions due to varying levels of endogenous SMAC/DIABLO expression and potential matrix effects.
A comprehensive validation strategy for SMAC/DIABLO antibodies should include multiple levels of controls. Positive controls should include cell lines known to express SMAC/DIABLO (such as SH-SY5Y, HeLa, Jurkat, HEK-293, and HepG2 cells). Recombinant SMAC/DIABLO protein serves as an essential positive control for Western blot analysis, establishing the correct molecular weight band pattern. Negative controls should include antibody omission, isotype controls, and ideally, SMAC/DIABLO-knockout or knockdown samples. For knockdown validation, siRNA treatment followed by Western blot analysis demonstrating reduced signal intensity provides strong evidence of antibody specificity. Pre-absorption of the antibody with immunizing peptide should eliminate specific staining. Cross-reactivity testing across species is advisable if working with non-human models, as anti-human SMAC/DIABLO antibodies have varying reactivity with mouse and rat homologs. Finally, dual-antibody validation using antibodies targeting different epitopes of SMAC/DIABLO provides robust confirmation of specificity when concordant results are obtained.
Investigating the temporal relationship between cytochrome c and SMAC/DIABLO release during apoptosis requires sophisticated time-course experiments with dual-labeling techniques. Implement live-cell imaging using fluorescently-tagged SMAC/DIABLO and cytochrome c constructs for real-time visualization, or employ fixed-cell immunofluorescence with antibodies against both proteins at precise time intervals following apoptotic stimulation. For biochemical analysis, perform subcellular fractionation at defined time points (e.g., 0, 15, 30, 60, 120, and 240 minutes post-stimulation) and analyze both cytosolic and mitochondrial fractions by Western blot. Research has indicated that while cytochrome c and SMAC/DIABLO are generally released with similar kinetics in response to death receptor ligation, subtle differences may exist depending on the apoptotic stimulus and cell type. Flow cytometry with intracellular staining for both proteins provides quantitative single-cell resolution data that can reveal population heterogeneity in release timing. For the most rigorous temporal analysis, combine these approaches with caspase activity assays to correlate mitochondrial protein release with downstream apoptotic events.
Distinguishing between different mechanisms of SMAC/DIABLO release requires multi-parametric experimental strategies. Begin with specific inhibitors targeting distinct release pathways: cyclosporin A for permeability transition pore (PTP) inhibition, Bcl-2 family modulators (ABT-737/ABT-263) for BAX/BAK-mediated outer membrane permeabilization, and caspase inhibitors (z-VAD-fmk) to test feedback amplification loops. Combine these pharmacological approaches with genetic manipulation using CRISPR/Cas9 knockout or siRNA knockdown of key regulatory components (BAX/BAK, VDAC, cyclophilin D). For direct assessment of mitochondrial permeability transition, measure mitochondrial membrane potential (ΔΨm) using potentiometric dyes (TMRE, JC-1) simultaneously with SMAC/DIABLO release. Employ super-resolution microscopy to visualize the formation of BAX/BAK pores in the outer mitochondrial membrane coincident with SMAC/DIABLO release. Electron microscopy can provide ultrastructural evidence of mitochondrial morphological changes during release events. Finally, in vitro reconstitution assays using isolated mitochondria exposed to recombinant proteins (tBID, BAX) or apoptotic stimuli allow controlled examination of release mechanisms under defined conditions.
SMAC/DIABLO antibodies serve as essential tools in researching SMAC mimetics and therapy resistance mechanisms in cancer. For evaluating SMAC mimetic efficacy, establish baseline endogenous SMAC/DIABLO expression levels across cancer cell lines using validated antibodies in Western blot and immunohistochemistry analyses, as expression levels correlate with therapeutic response. Employ co-immunoprecipitation with SMAC/DIABLO antibodies to assess displacement of endogenous SMAC/DIABLO from IAPs by mimetic compounds, confirming the mechanism of action. In therapy resistance studies, perform longitudinal analysis of SMAC/DIABLO expression and subcellular localization in sensitive versus resistant cell lines using immunofluorescence and fractionation approaches. Develop resistance models through chronic exposure to SMAC mimetics and use antibodies to track accompanying changes in SMAC/DIABLO-IAP interactions via proximity ligation assays. For translational research, conduct immunohistochemical analysis of patient biopsies before and after treatment to correlate SMAC/DIABLO expression patterns with clinical outcomes. Combine these approaches with quantitative proteomic analysis of SMAC-interacting proteins immunoprecipitated using validated antibodies to identify novel resistance mechanisms involving alterations in SMAC/DIABLO binding partners or post-translational modifications.
Inconsistent SMAC/DIABLO detection in Western blot analysis can stem from multiple factors requiring systematic troubleshooting. Protein degradation is a primary concern, particularly given SMAC/DIABLO's involvement in apoptotic pathways – always use fresh samples with complete protease inhibitor cocktails and maintain cold temperatures during preparation. Sample preparation artifacts may occur if mitochondrial integrity is compromised during processing, causing premature SMAC/DIABLO release into cytosolic fractions. Transfer efficiency issues are common with small proteins like mature SMAC/DIABLO (~21 kDa); optimize transfer conditions using low molecular weight-optimized protocols (higher methanol percentage, lower voltage, longer transfer time). Buffer composition significantly impacts detection – use reducing conditions (as indicated in validation data) and the appropriate immunoblot buffer group (Buffer Group 6 has been validated for some antibodies). The antibody concentration needs careful optimization; excessive dilution reduces sensitivity while over-concentration increases background. Clone-specific variations exist – monoclonal antibodies typically detect a strong band at approximately 20 kDa while some polyclonal preparations may detect additional bands. If experiencing inconsistent results, try stripping and re-probing membranes with alternative SMAC/DIABLO antibodies targeting different epitopes.
Addressing cross-reactivity challenges in multi-species SMAC/DIABLO studies requires rigorous validation and strategic experimental design. Begin by performing sequence alignment analysis between human, mouse, and rat SMAC/DIABLO homologs to identify regions of divergence that might affect antibody recognition. Test each antibody against recombinant proteins or lysates from all target species under identical conditions before proceeding with complex experiments. When absolute cross-reactivity confirmation is needed, use knockout or knockdown controls from each species. For immunohistochemistry applications, include absorption controls with species-specific blocking peptides to confirm specificity. If working with multiple species is essential but cross-reactivity is limited, consider using species-specific antibody pairs with validated epitope information. When quantitative comparisons across species are required, develop correction factors based on standard curves generated with recombinant proteins from each species. Finally, when reporting results, clearly document the validation steps performed and acknowledge any cross-reactivity limitations that might affect data interpretation. For critical applications, consider developing custom antibodies against highly conserved epitopes if commercially available options prove inadequate for multi-species detection.
Addressing molecular weight discrepancies in SMAC/DIABLO detection requires understanding the technical and biological factors influencing apparent protein size. The observed differences between Western blot detection at approximately 22 kDa and Simple Western techniques showing bands at approximately 29 kDa likely reflect technical variations in molecular weight determination methodologies rather than true biological differences. When encountering such discrepancies, researchers should first verify antibody specificity through knockout/knockdown controls. Next, directly compare detection methods using the same samples and antibody preparations to isolate methodological variables. Post-translational modifications can significantly impact migration patterns – phosphorylation, ubiquitination, or other modifications may be differentially preserved in various sample preparation methods. Verify the identity of ambiguous bands through mass spectrometry analysis of excised gel regions or immunoprecipitated protein. For quantitative studies, develop consistent recognition standards by always including recombinant SMAC/DIABLO protein as a migration reference. When reporting results with molecular weight variations, thoroughly document the exact experimental conditions, including gel percentage, running buffer composition, and molecular weight markers used. Finally, consider that splice variants or alternative processing may exist that are preferentially detected by different antibodies or under different experimental conditions.
Statistical analysis of SMAC/DIABLO translocation requires approaches tailored to the specific experimental design and data characteristics. For Western blot quantification of cytosolic versus mitochondrial fractions, paired statistical tests (paired t-tests or Wilcoxon signed-rank tests) are appropriate when comparing treatments within the same experimental preparation. Time-course translocation data should be analyzed using repeated measures ANOVA with appropriate post-hoc tests, accounting for the non-independence of sequential measurements. For immunofluorescence data quantifying subcellular localization, employ image analysis algorithms that objectively define mitochondrial and cytosolic regions based on mitochondrial marker co-staining, followed by intensity ratio calculations. When analyzing heterogeneous cell populations, consider single-cell analysis approaches that capture the distribution of responses rather than population averages. For dose-response relationships between apoptotic stimuli and SMAC/DIABLO translocation, nonlinear regression models should be applied to determine EC50 values and Hill coefficients. Survival analysis methodologies (Kaplan-Meier curves, Cox proportional hazards models) are valuable when correlating SMAC/DIABLO translocation timing with cellular outcomes. For complex experimental designs involving multiple variables (stimulus type, cell type, time, drug concentration), multivariate statistical approaches such as principal component analysis or partial least squares regression can identify key factors driving translocation dynamics.
Distinguishing specific from non-specific bands in complex tissue samples requires a systematic validation approach combining multiple controls and analytical techniques. Begin by establishing a reference profile using cell lines with confirmed SMAC/DIABLO expression (SH-SY5Y, HeLa, Jurkat, HEK-293) alongside recombinant protein standards. For each new tissue type, perform antibody titration experiments to identify the optimal concentration that maximizes specific signal while minimizing background. Include absorption controls where antibodies are pre-incubated with immunizing peptide – specific bands should disappear while non-specific signals persist. When analyzing tissues with complex protein composition, higher stringency washing conditions (increased salt concentration or detergent) may help eliminate non-specific binding. For definitive band identification, perform immunoprecipitation followed by mass spectrometry analysis of the enriched protein(s). Use siRNA knockdown in appropriate primary cell cultures derived from the tissue of interest to verify band specificity. When multiple antibodies are available, compare band patterns across antibodies targeting different SMAC/DIABLO epitopes – specific bands should be consistently detected. For challenging tissues with high background, consider alternative detection systems such as highly sensitive chemiluminescent substrates or fluorescent secondary antibodies with narrow emission spectra to improve signal-to-noise ratios. Always include appropriate loading controls optimized for the specific tissue type being analyzed.
SMAC/DIABLO (Second Mitochondria-derived Activator of Caspases/Direct IAP Binding Protein with Low pI) is a mitochondrial protein that plays a crucial role in the regulation of apoptosis. This protein is encoded by the DIABLO gene in humans and is known for its ability to bind and neutralize inhibitor of apoptosis proteins (IAPs), thereby promoting the activation of caspases and the apoptotic process .
SMAC/DIABLO is initially synthesized as a precursor protein with a mitochondrial targeting sequence (MTS) at its N-terminal. Upon import into the mitochondria, this sequence is cleaved off, resulting in the mature form of the protein. The mature SMAC/DIABLO protein is characterized by an N-terminal tetrapeptide sequence (Ala-Val-Pro-Ile) that is essential for its interaction with IAPs .
The primary function of SMAC/DIABLO is to promote apoptosis by binding to IAPs, such as XIAP (X-linked Inhibitor of Apoptosis Protein), and preventing them from inhibiting caspases. This interaction is crucial for the activation of caspases, which are the key executioners of apoptosis .
Apoptosis, or programmed cell death, is a vital process for maintaining cellular homeostasis and eliminating damaged or unwanted cells. SMAC/DIABLO is released from the mitochondria into the cytosol in response to apoptotic stimuli. Once in the cytosol, it binds to IAPs and neutralizes their inhibitory effect on caspases, thereby facilitating the apoptotic process .
Due to its pro-apoptotic function, SMAC/DIABLO is implicated in various types of cancer. The overexpression of IAPs in cancer cells can lead to resistance to apoptosis, contributing to tumor growth and survival. Therefore, SMAC/DIABLO and its mimetics are being explored as potential therapeutic agents to enhance the efficacy of cancer treatments by promoting apoptosis in cancer cells .
Mouse anti-human SMAC/DIABLO antibodies are monoclonal antibodies developed to specifically recognize and bind to the human SMAC/DIABLO protein. These antibodies are widely used in research to study the expression and function of SMAC/DIABLO in various biological contexts. They are also utilized in techniques such as Western blotting, immunohistochemistry, and immunoprecipitation to detect and quantify SMAC/DIABLO in different samples .