Phospho-Bad (Ser155) Antibody

Shipped with Ice Packs
In Stock

Description

Biological Context and Functional Mechanisms

BAD is a pro-apoptotic Bcl-2 family protein that promotes cell death by binding and neutralizing anti-apoptotic proteins like Bcl-XL. Phosphorylation at Ser155 inhibits BAD’s apoptotic function by:

  • Blocking its interaction with Bcl-XL

  • Faculating binding to 14-3-3 scaffold proteins, sequestering BAD in the cytoplasm

This phosphorylation event is regulated by kinases (e.g., AKT, PKA) and phosphatases (e.g., PP2C). Dysregulation of Ser155 phosphorylation is implicated in cancer progression and metabolic disorders .

Role in Cancer Development

  • Elevated pBAD (Ser155) in Cancer Cells: Comparative studies of immortalized normal vs. cancer cell lines (ovarian, colon, breast) revealed 2–3× higher pBAD (Ser155) levels in cancer cells. This correlates with reduced PP2C phosphatase expression in tumors, enhancing pro-survival signaling .

  • Chemoresistance: Ovarian cancers with low PP2C levels (resulting in sustained Ser155 phosphorylation) show poorer responses to platinum-based therapy .

Therapeutic Potential in Diabetes

  • β-Cell Protection: A phospho-mimetic BAD variant (S155D) protects pancreatic β-cells from apoptosis induced by cytokines, hypoxia, or ER stress. This effect requires glucokinase (GK) activation, highlighting a dual role in metabolism and survival .

  • Islet Transplantation: Pretreating donor islets with a BAD BH3 phospho-mimetic (S155D) improves engraftment and glycemic control in diabetic mice, demonstrating translational potential .

Key Applications in Experimental Research

  • Mechanistic Studies: Used to investigate BAD phosphorylation dynamics in apoptosis, chemoresistance, and metabolic regulation .

  • Diagnostic Development: Serves as a biomarker tool for assessing PP2C activity and BAD pathway integrity in cancer biopsies .

  • Therapeutic Screening: Facilitates testing of kinase inhibitors or phosphatase activators targeting BAD-mediated survival pathways .

Limitations and Considerations

  • Research Use Only (RUO): Not validated for diagnostic or therapeutic applications .

  • Context-Dependent Results: Phosphorylation status varies by tissue type and stress conditions, requiring rigorous controls in experimental design .

Product Specs

Form
Supplied at 1.0 mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150 mM NaCl, 0.02% sodium azide and 50% glycerol.
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method and location. Please consult your local distributors for specific delivery timelines.
Synonyms
Bad antibody; Bbc6Bcl2-associated agonist of cell death antibody; BAD antibody; Bcl-2-binding component 6 antibody; Bcl-xL/Bcl-2-associated death promoter antibody; Bcl2 antagonist of cell death antibody
Target Names
Bad
Uniprot No.

Target Background

Function
Phospho-Bad (Ser155) Antibody promotes cell death. It effectively competes for binding to Bcl-X(L), Bcl-2, and Bcl-W, influencing the level of heterodimerization of these proteins with BAX. It can reverse the death repressor activity of Bcl-X(L) but not that of Bcl-2. Phospho-Bad (Ser155) Antibody appears to act as a link between growth factor receptor signaling and apoptotic pathways.
Gene References Into Functions
  1. This study demonstrates, for the first time, that genetic knockout of Bad provides protection against epileptic seizures in Kcna1-/- mice, a genetic model of epilepsy characterized by sudden unexplained death. PMID: 29171006
  2. BAD knockout reduced epileptiform activity, and this effect was lost upon knockout or pharmacological inhibition of KATP channels. PMID: 29368690
  3. Bad is not essential for TNF-mediated cell death. PMID: 25611386
  4. Results suggest that the regulation of the pro-apoptotic activity of BAD plays a crucial role in the pathogenic mechanisms leading to primary pigmented nodular adrenocortical disease tumor formation. PMID: 24865460
  5. Fasting may increase the uptake of beta-hydroxybutyrate by decreasing BAD in the brain during hypoglycemia. PMID: 25043191
  6. Results indicate the downstream targets of insulin, cyclin D1, BAD, alpha-MHC, and GATA-4, elucidating a molecular mechanism by which insulin promotes cell proliferation and differentiation. PMID: 24020834
  7. Our study suggests that Bad and Bmf co-regulate lymphocyte homeostasis and limit spontaneous transformation through mechanisms that may not be exclusively linked to the induction of lymphocyte apoptosis. PMID: 22430207
  8. Results reveal that IKK inhibits TNFalpha-induced apoptosis through two distinct yet cooperative mechanisms: activation of the survival factor NF-kappaB and inactivation of the pro-apoptotic BH3-only BAD protein. PMID: 23332762
  9. RNAi-mediated silencing of STAT1 in soft tissue sarcoma (STS) cells was sufficient to increase expression of the apoptotic mediators Fas and Bad and to enhance the sensitivity of STS cells to Fas-mediated apoptosis. PMID: 22805310
  10. BAD modulates counterregulatory responses to hypoglycemia and protective glucoprivic feeding. PMID: 22162752
  11. The regulation of BAD by uremic toxins and the sensitization of vascular smooth muscle cells to apoptosis upon BAD induction were explored. PMID: 22172950
  12. Tonicity-induced COX-2 expression and PGE2 synthesis in the renal medulla involves phosphorylation and inactivation of the pro-apoptotic protein Bad, thereby counteracting apoptosis in renal medullary epithelial cells. PMID: 21716255
  13. Caspase-3 is activated by the BAD-BAX cascade, resulting in long-term depression induction in the hippocampus. PMID: 21609830
  14. JNK1 is required for erythropoietin-mediated cell survival through phosphorylation and inactivation of the pro-apoptotic, Bcl-2 homology domain 3 (BH3)-only Bcl-associated death protein (Bad). PMID: 21095239
  15. Bad protein cooperates with Bim protein in certain apoptotic responses and in the suppression of g-irradiation-induced thymic lymphoma. PMID: 20431598
  16. Data show that loss of Bmf reduced the pressure to inactivate p53, whereas Bad deficiency did not, identifying Bmf as a novel component of the p53-independent tumor suppressor pathway triggered by c-Myc. PMID: 19965635
  17. The beta-arrestin 1-dependent ERK1/2 activation engaged by GLP-1 mediates the Ser-112 phosphorylation of Bad. PMID: 19915011
  18. The interaction of Bad with lipid rafts is a dynamic process regulated by IL-4 and involved in the control of apoptosis. PMID: 11907096
  19. Activation by therapeutic inhibition of epidermal growth factor receptor and transactivation by insulin-like growth factor receptor. PMID: 12011069
  20. Bcl-x(L) and Bcl-w target protein phosphatase 1alpha to Bad. PMID: 12115603
  21. Phosphorylation at serine 128 by activation of the JNK signaling pathway. PMID: 12189144
  22. BAD phosphorylation protects cells from the detrimental effects of apoptotic stimuli and attenuates death pathway signaling by increasing the threshold at which mitochondria release cytochrome c to induce cell death. PMID: 12431371
  23. Bad apoptotic protein alone or in combination with Bax apoptotic protein and the prostatic-specific promoter ARR(2)PB was an effective therapy for experimental prostatic neoplasms. PMID: 12490000
  24. Candida albicans phospholipomannan promotes survival of phagocytosed yeasts through modulation of this protein's phosphorylation and macrophage apoptosis. PMID: 12551950
  25. HSV-1 US3 protein kinase blocks the caspases that cleave BAD at either residue 56 or 61, predicted to render the protein more pro-apoptotic, or at residue 156, which would inactivate the protein. PMID: 12743316
  26. Pro-apoptotic BAD suppresses tumorigenesis in the lymphocyte lineage. PMID: 12876200
  27. A combination of proteomics, genetics, and physiology indicates an unexpected role for BAD in integrating pathways of glucose metabolism and apoptosis. PMID: 12931191
  28. PP2A dephosphorylation of pSer112 is the key initiating event regulating the activation of BAD during interleukin-3 withdrawal-induced apoptosis. PMID: 12944463
  29. BAD is a substrate for pim-2 oncogene proto-oncogene. PMID: 12954615
  30. Regulation of Bad phosphorylation plays an active role in mediating anti-IgM-induced apoptosis of immature B cells. PMID: 14585539
  31. JNK is required for IL-3-mediated cell survival through phosphorylation and inactivation of the pro-apoptotic Bcl-2 family protein BAD. PMID: 14967141
  32. Data show that the Bcl-2 homology 3 domain-only protein, Bad, is involved in cell death following IL-7 withdrawal from D1 cells, an IL-7-dependent murine thymocyte cell line. PMID: 15123689
  33. Mechanisms that regulate the conversion of BAD from an anti-death to a pro-death factor include alternative splicing that produces N-terminally truncated BAD(S) and conversion by caspases into a pro-death fragment that resembles the short splice variant. PMID: 15231831
  34. Alteration of lipid rafts is an early event in the apoptotic cascade indirectly induced by interleukin-4 deprivation via PP1alpha activation, dephosphorylation of cytoplasmic Bad, and caspase activation. PMID: 15634756
  35. Bad phosphorylation is not essential for PKB-mediated survival signaling in hemopoietic cells. PMID: 15843895
  36. Pak1-dependent Raf-1 phosphorylation regulates its mitochondrial localization, phosphorylation of BAD, and Bcl-2 association. PMID: 15849194
  37. BAD induces apoptosis upon detecting the coincidence of G2/M phase and growth factor deprivation. PMID: 15901741
  38. Phosphorylation of BAD Serine 128 exerts cell-specific effects on apoptosis. PMID: 15907327
  39. All three Pim kinase family members predominantly phosphorylate Bad on Ser112 and, in addition, are capable of phosphorylating Bad on multiple sites associated with the inhibition of the pro-apoptotic function of Bad in HEK-293 cells. PMID: 16403219
  40. Cellular cholesterol biosynthesis is critical for the activation and maintenance of the Akt-Bad cell survival cascade in response to growth factors such as insulin. PMID: 16513830
  41. These data establish a connection between calcium overload and mitochondria-mediated death pathways in outer hair cells and also suggest a dual role for BAD. PMID: 16521126
  42. The interaction of BAD with membranes is tied to binding of 14-3-3 protein and activation and membrane translocation of Bcl-XL. PMID: 16603546
  43. Study shows, using spectroscopic methods, that the BH3-only proteins Bim, Bad, and Bmf are unstructured in the absence of binding partners. PMID: 16645638
  44. Bad was not required for cell death following IL-3 withdrawal, suggesting changes to phosphorylation of Bad play only a minor role in apoptosis. PMID: 16705087
  45. Both gonadotropin-releasing hormone and epidermal growth factor (EGF) caused rapid phosphorylation of BAD. PMID: 16741954
  46. The pro-apoptotic protein Bad is a key player in cell survival decisions and is regulated post-translationally by several signaling networks. PMID: 17535812
  47. Raf-1 in beta-cells led to a striking loss of Bad phosphorylation at serine 112 and an increase in the protein levels of both Bad and Bax. PMID: 18006502
  48. These findings provide genetic proof of the bifunctional activities of BAD in both beta cell survival and insulin secretion. PMID: 18223655
  49. Thr-201 phosphorylation of Bad by JNK1 is required for PFK-1 activation. PMID: 18469002
  50. BAD is the only BCL-2 family protein expressed in parietal cells. PMID: 18779780

Show More

Hide All

Database Links

KEGG: mmu:12015

STRING: 10090.ENSMUSP00000025910

UniGene: Mm.4387

Protein Families
Bcl-2 family
Subcellular Location
Mitochondrion outer membrane. Cytoplasm.

Q&A

What is the biological significance of BAD phosphorylation at Ser155?

Phosphorylation at Ser155 in the BH3 domain of BAD by PKA plays a critical role in blocking the dimerization of BAD and Bcl-xL . This phosphorylation event is a key regulatory mechanism that inhibits BAD's pro-apoptotic function, promoting cell survival. When phosphorylated at Ser155, BAD cannot engage and neutralize pro-survival BCL-2 family proteins, thus preventing the initiation of apoptosis . This phosphorylation site represents a distinct regulatory mechanism from other well-characterized phosphorylation sites (S112, S136) and has specific implications for cellular metabolism and survival signaling pathways.

What applications are Phospho-Bad (Ser155) antibodies validated for?

Most commercially available Phospho-Bad (Ser155) antibodies have been validated for multiple applications:

ApplicationTypical Dilution RangeNotes
Western Blotting1:500-1:2000Primary detection method
Immunohistochemistry1:100-1:300For tissue sections
Immunofluorescence1:200-1:1000For cellular localization studies
ELISA1:10000For quantitative analysis

Researchers should note that optimal dilutions may vary between different antibody sources and experimental conditions. Validation with appropriate controls is always recommended before proceeding with experimental applications .

How specific are Phospho-Bad (Ser155) antibodies to the phosphorylated form?

Phospho-Bad (Ser155) antibodies are designed to be highly specific for the phosphorylated form of BAD at Ser155. These antibodies are typically produced by immunizing rabbits with synthetic phosphopeptides corresponding to the region surrounding Ser155 and then purified using affinity chromatography with epitope-specific phosphopeptides . Non-phospho specific antibodies are removed during purification by chromatography using non-phosphopeptides . Cross-reactivity testing indicates that quality antibodies from reputable sources detect BAD only when phosphorylated at Ser155 and do not cross-react with other phosphorylation sites (such as S112 or S136) or with non-phosphorylated BAD .

What species reactivity is expected for Phospho-Bad (Ser155) antibodies?

Most commercially available Phospho-Bad (Ser155) antibodies demonstrate reactivity with multiple species:

SpeciesReactivityNotes
HumanConfirmedMost commonly tested
MouseConfirmedOften used in β-cell research models
RatConfirmedValidated in several antibody preparations

Some antibodies share 100% sequence homology with additional species but may not have been specifically tested for reactivity. Researchers should consult the manufacturer's specifications for confirmation of cross-reactivity with their specific experimental model .

How can I distinguish between the effects of phosphorylation at Ser155 versus other phosphorylation sites (S112, S136) in BAD-mediated cellular processes?

Distinguishing between the effects of different phosphorylation sites requires a multi-faceted approach:

  • Use site-specific phospho-antibodies: Employ antibodies that specifically recognize phosphorylation at S155, S112, or S136 in parallel experiments.

  • Phospho-mimetic mutants: Utilize BAD mutants like S155D (mimicking phosphorylation) and compare with other phospho-mimetics (S112D, S136D) or non-phosphorylatable mutants (S155A) .

  • Functional readouts: BAD phosphorylation at S155 specifically blocks dimerization with Bcl-xL, while S112 and S136 phosphorylation promotes binding to 14-3-3 proteins .

  • Kinase inhibition: PKA specifically phosphorylates S155, while other kinases like AKT target S136 and p90RSK targets S112. Using specific kinase inhibitors can help delineate phosphorylation events .

  • Metabolism connection: S155 phosphorylation uniquely connects to glucose metabolism through glucokinase (GK) binding, which doesn't occur with other phosphorylation sites .

Research by Danial et al. has demonstrated that BAD S155D and BAD AAA (L151A, S155A, D156A) mutants can be particularly useful in distinguishing the metabolic versus apoptotic functions, as both block pro-apoptotic activity but only S155D activates glucokinase .

What methodological considerations should be taken into account when using Phospho-Bad (Ser155) antibodies in western blotting experiments?

Several critical methodological considerations must be addressed when using Phospho-Bad (Ser155) antibodies for western blotting:

  • Sample preparation:

    • Use phosphatase inhibitors in lysis buffers to prevent dephosphorylation

    • Process samples quickly and maintain cold temperatures

    • Consider using stimuli known to induce S155 phosphorylation (PKA activators)

  • Controls:

    • Include positive controls (cells with known BAD S155 phosphorylation)

    • Use phosphatase-treated samples as negative controls

    • Consider including BAD knockout or knockdown samples

  • Protocol optimization:

    • Molecular weight: BAD typically runs at approximately 23 kDa

    • Optimize transfer conditions for small proteins

    • Blocking solutions should avoid phospho-epitope masking (use BSA rather than milk)

    • Dilution range: 1:500-1:2000 depending on antibody source

  • Sensitivity considerations:

    • Some antibodies detect only transfected levels of phospho-BAD

    • Endogenous detection may require enrichment by immunoprecipitation

  • Data interpretation:

    • Confirm specificity with peptide competition assays

    • Consider parallel blotting with total BAD antibody

    • Quantify phospho-BAD/total BAD ratio for accurate assessment

How do experimental conditions affect the detection of phosphorylated BAD at Ser155 in cell and tissue samples?

The detection of phosphorylated BAD at Ser155 is highly sensitive to experimental conditions:

  • Cell stimulation conditions:

    • PKA activation is the primary driver of S155 phosphorylation

    • cAMP-elevating agents (forskolin, IBMX) enhance phosphorylation

    • Glucose metabolism can influence phosphorylation states

    • Stress conditions (cytokines, oxidative stress) may alter phosphorylation patterns

  • Timing considerations:

    • Phosphorylation is a dynamic event; optimal detection time points must be determined

    • Survival stimuli temporarily enhance phosphorylation before returning to baseline

    • In β cells, timing relative to metabolic stress is critical

  • Tissue-specific considerations:

    • BAD phosphorylation patterns differ between tissues (pancreatic β cells vs. neurons)

    • Tissue fixation methods can affect phospho-epitope preservation

    • Fresh frozen tissues may preserve phosphorylation better than fixed specimens

  • Disease states:

    • Altered phosphorylation patterns occur in pathological conditions

    • Rapamycin treatment enhances phosphorylation at S112/S136 but not S155

    • Diabetes models show distinct alterations in BAD phosphorylation patterns

Research by Danial et al. demonstrated that β cells under inflammatory cytokine stress, oxidative stress, or ER stress show distinct patterns of BAD phosphorylation that can be modulated by phospho-mimetic interventions .

What are the technical challenges in accurately distinguishing between BAD phosphorylation states in multi-protein complexes?

Distinguishing BAD phosphorylation states in multi-protein complexes presents several technical challenges:

  • Complex-dependent epitope accessibility:

    • BAD phosphorylation at S155 affects its binding to Bcl-xL, potentially masking epitopes

    • 14-3-3 binding to BAD (driven by S112/S136 phosphorylation) may sterically hinder antibody access to S155

    • Protein complexes may need to be disrupted before immunodetection

  • Co-immunoprecipitation considerations:

    • Choice of antibody for immunoprecipitation may bias complex recovery

    • Sequential immunoprecipitation might be necessary to distinguish subcomplexes

    • Native vs. denaturing conditions affect complex stability

  • Quantitative analysis challenges:

    • Phosphorylation at multiple sites occurs simultaneously

    • Stoichiometry of phosphorylation varies by site and condition

    • Quantifying multiple phosphorylation states requires multiplexed approaches

  • Subcellular localization:

    • Phospho-BAD distributes differently between cytosol and mitochondria

    • Rapamycin treatment promotes cytosolic accumulation of phospho-BAD

    • Fractionation protocols must preserve phosphorylation status

  • Advanced approaches:

    • Proximity ligation assays can detect specific phospho-BAD/protein interactions

    • Mass spectrometry can quantify multiple phosphorylation sites simultaneously

    • FRET-based approaches can monitor dynamic changes in protein-protein interactions

Research by Jin et al. showed that rapamycin promotes BAD accumulation in the cytosol, enhances BAD/14-3-3 interaction, and reduces BAD/Bcl-XL binding, highlighting the dynamic nature of these complexes .

How can Phospho-Bad (Ser155) antibodies be integrated into broader studies of apoptosis and cell survival pathways?

Integration of Phospho-Bad (Ser155) antibodies into comprehensive apoptosis and survival studies requires:

  • Pathway analysis integration:

    • Combine with analyses of upstream regulators (PKA, cAMP signaling)

    • Correlate with metabolic pathway activation (glucokinase activity)

    • Evaluate downstream mitochondrial integrity markers (cytochrome c release, membrane potential)

  • Multi-parameter experimental design:

    • Combine phospho-Bad detection with apoptosis assays (Annexin V, TUNEL)

    • Monitor mitochondrial function in parallel (respiration, membrane potential)

    • Track metabolic parameters (glucose utilization, ATP production)

  • Therapeutic intervention studies:

    • Use BAD BH3 mimetics as experimental interventions (SAHB peptides)

    • Evaluate effects of kinase inhibitors on phosphorylation patterns

    • Study phospho-BAD modulation by drugs like rapamycin

  • Disease model applications:

    • Apply to models of diabetes for β cell survival assessment

    • Evaluate in cancer models for chemoresistance mechanisms

    • Study neurodegenerative conditions where apoptosis regulation is dysregulated

  • Technological integration:

    • Combine with live-cell imaging of apoptosis markers

    • Integrate with phospho-proteomics for broader pathway analysis

    • Apply in high-content screening approaches

Research by Danial et al. demonstrated that pharmacologic mimetics of phosphorylated BAD BH3 domain (SAHB peptides) provide β cell protection and represent a novel therapeutic approach, highlighting how phospho-BAD research translates to intervention strategies .

What are the optimal fixation and extraction protocols for preserving Phospho-Bad (Ser155) epitopes in different sample types?

Optimal protocols for preserving Phospho-Bad (Ser155) epitopes vary by sample type:

  • Cell culture samples:

    • Preferred fixation: 4% paraformaldehyde (10-15 minutes at room temperature)

    • Immediate extraction in phosphatase inhibitor-containing buffers

    • Avoid methanol fixation which can extract phospholipids and associated proteins

    • For suspension cells: Consider poly-L-lysine coating before fixation

  • Tissue samples:

    • Fresh frozen sections provide superior phospho-epitope preservation

    • If paraffin embedding is necessary, use phosphate-buffered formalin

    • Antigen retrieval: Citrate buffer (pH 6.0) is typically effective

    • Post-fixation washing should include phosphatase inhibitors

  • Extraction considerations:

    • Lysis buffer composition: RIPA or NP-40 buffers with phosphatase inhibitors

    • Include both serine/threonine and tyrosine phosphatase inhibitors

    • Maintain cold temperatures throughout processing

    • Sonication may improve extraction of membrane-associated complexes

  • Special sample types:

    • Pancreatic islets: Require gentle dissociation before processing

    • Brain tissue: Rapid processing critical due to high phosphatase activity

    • Tumor samples: Consider tumor heterogeneity in sampling

Empirical testing of multiple fixation conditions may be necessary for optimal results with specific tissue types and antibody sources.

How can I troubleshoot inconsistent results when using Phospho-Bad (Ser155) antibodies across different experimental systems?

Troubleshooting inconsistent results requires systematic evaluation of multiple variables:

  • Antibody-specific factors:

    • Verify antibody lot consistency and storage conditions

    • Test multiple antibody sources/clones

    • Determine optimal concentration for each system

    • Consider antibody validation with peptide competition

  • Sample preparation variables:

    • Standardize cell culture conditions (passage number, confluency)

    • Use consistent stimulation protocols and timing

    • Ensure phosphatase inhibitor effectiveness

    • Standardize protein quantification methods

  • Technical execution:

    • Optimize blocking conditions (BSA vs. milk proteins)

    • Adjust antibody incubation time and temperature

    • Implement consistent washing protocols

    • Consider automated systems for improved reproducibility

  • System-specific optimization:

    • Different cell lines may require adjusted lysis conditions

    • Primary cells vs. cell lines have different baseline phosphorylation

    • Tissue-specific protocols may need adaptation

  • Controls and standards:

    • Include positive controls (PKA-activated samples)

    • Use phosphatase-treated negative controls

    • Consider phospho-peptide standards for quantitative work

    • Include internal loading controls for normalization

Systematic documentation of all variables and conditions can help identify the sources of inconsistency across experimental systems.

What methods can be used to quantitatively assess the stoichiometry of BAD phosphorylation at Ser155 versus other phosphorylation sites?

Quantitative assessment of BAD phosphorylation stoichiometry requires sophisticated approaches:

  • Mass spectrometry-based methods:

    • Targeted MS/MS for specific phosphopeptides

    • Parallel reaction monitoring for quantitative comparison

    • SILAC or TMT labeling for relative quantification

    • Absolute quantification using isotope-labeled standards

  • Antibody-based quantitative approaches:

    • Quantitative western blotting with site-specific antibodies

    • Normalization to total BAD protein

    • ELISA-based quantification of multiple phospho-sites

    • Multiplexed detection systems (Luminex, protein simple)

  • Mobility shift analysis:

    • Phos-tag SDS-PAGE to separate phosphorylated species

    • 2D gel electrophoresis to resolve multiple phospho-forms

    • Correlation of shifts with site-specific antibody detection

  • Genetic approaches for calibration:

    • Phospho-mimetic standards (S155D, S112D, S136D)

    • Non-phosphorylatable mutants as negative controls

    • Single, double, and triple phospho-site mutants for comparison

  • Mathematical modeling:

    • Kinetic models of multiple phosphorylation events

    • Bayesian approaches to infer phosphorylation stoichiometry

    • Integration of multiple data types for comprehensive assessment

Research by Jin et al. demonstrated that rapamycin treatment produced differential effects on BAD phosphorylation sites (enhancing S112/S136 but not S155), highlighting the importance of site-specific quantitative assessment .

How do I design validation experiments to confirm the specificity of Phospho-Bad (Ser155) antibody detection in my experimental system?

Comprehensive validation of Phospho-Bad (Ser155) antibody specificity requires multiple approaches:

  • Genetic validation:

    • Use BAD knockout cells/tissues as negative controls

    • Test with S155A mutant (non-phosphorylatable) vs. wild-type BAD

    • Compare with S155D phospho-mimetic positive control

    • Employ siRNA knockdown with reconstitution experiments

  • Biochemical validation:

    • Phosphatase treatment of samples to eliminate signal

    • Peptide competition with phospho and non-phospho peptides

    • Immunoprecipitation followed by mass spectrometry confirmation

    • Sequential probing with multiple phospho-specific antibodies

  • Pharmacological validation:

    • PKA activators should enhance S155 phosphorylation

    • PKA inhibitors should reduce signal

    • Rapamycin treatment should not affect S155 phosphorylation

    • Compare with known modulators of S112/S136 phosphorylation

  • Cross-reactivity assessment:

    • Test antibody against related proteins (other BCL-2 family members)

    • Evaluate reactivity with other phosphorylation sites (pS112, pS136)

    • Cross-check with multiple antibodies from different vendors

    • Consider species-specific variations in the epitope region

  • Technical controls:

    • Include isotype control antibodies

    • Test secondary antibody alone to exclude non-specific binding

    • Include gradient of antigen concentration to assess linearity

    • Perform western blot alongside functional assays to correlate results

Thorough validation ensures reliable interpretation of experimental results and should be documented in publications using these antibodies.

How can Phospho-Bad (Ser155) antibodies be used to study the intersection of metabolism and apoptosis in pancreatic β cells?

Phospho-Bad (Ser155) antibodies offer unique insights into metabolism-apoptosis crosstalk in β cells:

  • Metabolic stress models:

    • Monitor phospho-BAD changes during cytokine stress, oxidative stress, and ER stress

    • Correlate with glucokinase activity and glucose metabolism

    • Track changes during hypoxia and nutrient deprivation

    • Assess in diabetes models with progressive β cell dysfunction

  • Mitochondrial function analysis:

    • Co-localization studies of phospho-BAD with mitochondrial markers

    • Correlation with mitochondrial membrane potential

    • Integration with measurements of mitochondrial respiration

    • Assessment of glucose-stimulated insulin secretion pathways

  • Intervention studies:

    • Effects of phospho-BAD BH3 mimetics (SAHB peptides) on β cell survival

    • Glucokinase activator effects on BAD phosphorylation

    • PKA modulator impacts on the BAD-glucokinase axis

    • Anti-diabetic drug effects on BAD phosphorylation patterns

  • Translational applications:

    • Human islet studies comparing healthy vs. diabetic donors

    • Correlation with markers of β cell stress in patient samples

    • Therapeutic target identification based on phosphorylation patterns

    • Biomarker development for β cell stress states

Research by Danial et al. demonstrated that mimicking phosphorylated BAD at S155 protects β cells from multiple stress stimuli relevant to type 1 diabetes, highlighting the critical role of this phosphorylation site in β cell survival mechanisms .

What role does BAD phosphorylation at Ser155 play in cancer drug resistance, and how can this be studied using phospho-specific antibodies?

BAD phosphorylation at Ser155 has important implications for cancer drug resistance:

  • Resistance mechanism characterization:

    • Compare phospho-BAD (S155) levels in sensitive vs. resistant cancer cells

    • Assess changes during acquired resistance development

    • Correlate with expression of pro-survival BCL-2 family proteins

    • Study in relation to other BAD phosphorylation sites (S112, S136)

  • Drug response studies:

    • Monitor phospho-BAD dynamics during chemotherapy treatment

    • Assess impact of kinase inhibitors on BAD phosphorylation patterns

    • Study rapamycin effects on differential BAD phosphorylation sites

    • Correlate phosphorylation changes with apoptotic resistance

  • Therapeutic targeting approaches:

    • Test BAD BH3 mimetics to overcome resistance

    • Evaluate combination therapies targeting multiple phosphorylation sites

    • Study PKA inhibition as a sensitization strategy

    • Target metabolic vulnerabilities linked to BAD phosphorylation

  • Clinical correlations:

    • Phospho-BAD immunohistochemistry in patient samples

    • Correlation with treatment response and patient outcomes

    • Potential biomarker development for therapy selection

    • Analysis in circulating tumor cells or liquid biopsies

Research by Jin et al. demonstrated that rapamycin treatment enhances phosphorylation of BAD at S112 and S136 but not S155, contributing to rapamycin resistance. Simultaneous blockage of S112 and S136 phosphorylation significantly enhanced rapamycin sensitivity, highlighting the distinct roles of different phosphorylation sites in drug resistance .

How can multiplexed detection of multiple BAD phosphorylation sites provide insights into signaling network dynamics?

Multiplexed detection of BAD phosphorylation sites reveals complex signaling dynamics:

  • Multi-parametric analytical approaches:

    • Simultaneous detection of pS155, pS112, and pS136

    • Correlation with upstream kinase activities (PKA, Akt, p90RSK)

    • Integration with other BCL-2 family protein modifications

    • Inclusion of 14-3-3 binding and subcellular localization markers

  • Temporal dynamics analysis:

    • Time-course studies of phosphorylation site changes

    • Order of phosphorylation events following stimulation

    • Persistence of different phosphorylation states

    • Recovery dynamics after stress removal

  • Technological approaches:

    • Multiplex phospho-flow cytometry

    • Sequential reprobing of western blots

    • Multiplexed immunofluorescence imaging

    • Mass cytometry (CyTOF) for single-cell resolution

  • Network modeling:

    • Mathematical modeling of kinase-phosphatase networks

    • Prediction of phosphorylation site interdependencies

    • Feedback loop identification in BAD regulation

    • Integration with broader cell death/survival pathway models

  • Perturbation studies:

    • Systematic kinase inhibitor treatments

    • Genetic manipulation of upstream regulators

    • Metabolic pathway modulation

    • Growth factor withdrawal or stimulation

Research comparing rapamycin effects on different BAD phosphorylation sites demonstrated how integrating multiple phosphorylation measurements provides mechanistic insights into drug resistance that would be missed by single-site analysis .

What are the most effective imaging approaches for studying the subcellular localization and dynamics of phosphorylated BAD at Ser155?

Effective imaging of phosphorylated BAD at Ser155 requires specialized approaches:

  • Advanced immunofluorescence techniques:

    • Super-resolution microscopy (STED, STORM, SIM) for detailed localization

    • Optimized fixation protocols to preserve phospho-epitopes

    • Multiplexed detection with mitochondrial and cytosolic markers

    • Z-stack confocal imaging for 3D distribution

  • Live-cell imaging strategies:

    • FRET-based sensors for BAD phosphorylation dynamics

    • Split-GFP complementation for protein-protein interactions

    • Photoactivatable or photoconvertible BAD fusion proteins

    • Correlative light-electron microscopy for ultrastructural context

  • Proximity-based detection methods:

    • Proximity ligation assay for phospho-BAD/Bcl-xL interactions

    • BioID or APEX2 proximity labeling with phospho-mutants

    • FRAP (Fluorescence Recovery After Photobleaching) for mobility analysis

    • Optogenetic approaches to spatially control BAD phosphorylation

  • Quantitative image analysis:

    • Colocalization coefficients with mitochondrial markers

    • Intensity correlation analysis for protein interactions

    • Single-molecule tracking of phospho-BAD dynamics

    • Machine learning approaches for pattern recognition

  • Model systems:

    • Primary cell cultures with physiological expression levels

    • 3D organoid systems for tissue context

    • Tissue clearing techniques for intact organ imaging

    • In vivo imaging in transparent model organisms

Research has demonstrated that phosphorylation at S155 specifically affects BAD's association with mitochondria and interaction with Bcl-xL, making subcellular localization studies particularly informative for understanding its function .

How can computational modeling integrate phospho-specific BAD data to predict cellular outcomes in complex disease models?

Computational modeling with phospho-BAD data enables predictive understanding:

  • Multi-scale modeling approaches:

    • Molecular dynamics simulations of phosphorylation effects on BAD structure

    • Kinetic models of phosphorylation/dephosphorylation cycles

    • Agent-based models of mitochondrial membrane permeabilization

    • Population-level models of cell fate decisions

  • Data integration strategies:

    • Bayesian networks incorporating multiple phosphorylation sites

    • Machine learning classification of cell survival probability

    • Principal component analysis of phosphorylation patterns

    • Time-series analysis of phosphorylation dynamics

  • Disease-specific applications:

    • Diabetes models predicting β cell survival under stress conditions

    • Cancer models of treatment resistance mechanisms

    • Neurodegenerative disease progression models

    • Ischemia-reperfusion injury response prediction

  • Predictive capabilities:

    • Identification of critical phosphorylation thresholds for cell survival

    • Prediction of drug combination efficacy

    • Patient-specific response modeling from biopsy data

    • Optimal intervention timing based on phosphorylation dynamics

  • Validation approaches:

    • Experimental testing of model-derived hypotheses

    • Sensitivity analysis to identify key parameters

    • Comparison with clinical outcomes for validation

    • Iterative refinement based on new experimental data

Research by Danial et al. demonstrated that phospho-BAD status could predict β cell survival under various stress conditions, providing a foundation for computational models integrating phosphorylation data with cellular outcomes .

What are the key limitations in current Phospho-Bad (Ser155) antibody technology that researchers should be aware of?

Current phospho-BAD (Ser155) antibody technology has several important limitations:

  • Detection sensitivity challenges:

    • Many antibodies detect only transfected levels of phospho-BAD

    • Endogenous detection often requires enrichment techniques

    • Signal-to-noise ratio can be problematic in tissues with low expression

    • Quantitative linearity may be limited across concentration ranges

  • Specificity considerations:

    • Cross-reactivity with other phosphorylation sites may occur

    • Batch-to-batch variability affects reproducibility

    • Epitope masking in protein complexes limits detection

    • Context-dependent performance (fixed vs. frozen samples)

  • Temporal limitations:

    • Static measurements miss dynamic phosphorylation changes

    • Optimal fixation timing is critical but often difficult to standardize

    • Rapid dephosphorylation during sample processing

    • Half-life of phosphorylated BAD varies with experimental conditions

  • Technical constraints:

    • Limited multiplexing capability with other phosphorylation sites

    • Incompatibility with certain fixatives or buffer systems

    • Species-specific performance differences

    • Limited dynamic range for quantitative applications

  • Validation challenges:

    • Lack of standardized validation protocols across the field

    • Limited availability of appropriate knockout controls

    • Incomplete characterization of cross-reactivity profiles

    • Few studies directly comparing antibodies from different sources

Researchers should acknowledge these limitations in experimental design and data interpretation, implementing appropriate controls and validation steps for their specific experimental systems.

How should researchers interpret contradictory results between Phospho-Bad (Ser155) antibody detection and functional outcomes in cell survival assays?

Resolving contradictions between phospho-detection and functional outcomes requires systematic analysis:

  • Mechanistic considerations:

    • S155 phosphorylation is necessary but may not be sufficient for survival

    • Multiple phosphorylation sites act in concert (S112, S136, S155)

    • Threshold effects may exist in phosphorylation-function relationships

    • Timing disparities between phosphorylation and functional outcomes

  • Technical reconciliation approaches:

    • Confirm antibody specificity with appropriate controls

    • Validate functional assays with positive/negative controls

    • Perform time-course studies to align temporal relationships

    • Use genetic approaches (phospho-mimetics) to confirm causality

  • Contextual factors:

    • Cell type-specific differences in BAD regulation networks

    • Variations in BCL-2 family protein expression levels

    • Metabolic state influences on BAD function

    • Stress type and intensity affecting outcome interpretation

  • Quantitative considerations:

    • Phosphorylation stoichiometry may be critical (partial vs. complete)

    • Threshold levels of phospho-BAD required for functional outcomes

    • Subcellular localization affecting functional impact

    • Protein complex formation influencing detection vs. function

  • Experimental design improvements:

    • Include multiple methodologies for phosphorylation detection

    • Correlate with direct measurements of BAD-protein interactions

    • Implement dose-response designs for intervention studies

    • Integrate with broader signaling pathway analysis

Research by Danial et al. showed that BAD S155D and BAD AAA had similar effects on blocking apoptotic function but divergent effects on cell survival during stress, highlighting how functional outcomes depend on more than simply blocking BAD's pro-apoptotic function .

What are the emerging technologies that may overcome current limitations in studying BAD phosphorylation dynamics?

Emerging technologies promise to address current limitations in phospho-BAD research:

  • Advanced protein engineering approaches:

    • Genetically encoded FRET-based phosphorylation sensors

    • Split fluorescent protein complementation systems

    • Phosphorylation-dependent protein switches

    • Optogenetic control of BAD phosphorylation states

  • Next-generation antibody technologies:

    • Single-domain antibodies (nanobodies) for improved access to complexes

    • Synthetic recombinant antibodies with enhanced specificity

    • Aptamer-based detection of phosphorylation states

    • Affimers and other non-antibody binding scaffolds

  • Mass spectrometry innovations:

    • Top-down proteomics for intact protein analysis

    • Single-cell phosphoproteomics

    • Ion mobility separation for improved phospho-isomer discrimination

    • Targeted SWATH-MS for improved quantification

  • Spatial biology approaches:

    • Spatial transcriptomics integrated with phosphoprotein detection

    • Multiplexed ion beam imaging (MIBI) for tissue analysis

    • Digital spatial profiling of phosphoproteins

    • In situ proximity ligation with spatial resolution

  • Computational methods:

    • Deep learning for image analysis and pattern recognition

    • Multi-omics data integration frameworks

    • Network inference algorithms for phosphorylation cascades

    • Causal inference methods for mechanistic relationships

These emerging technologies will enable more dynamic, quantitative, and comprehensive analysis of BAD phosphorylation in physiological and pathological contexts.

What are the most critical considerations when translating findings from animal models to human studies regarding BAD phosphorylation at Ser155?

Critical considerations for translational research on BAD phosphorylation include:

  • Species-specific differences:

    • Sequence variations in BAD and regulatory proteins

    • Different tissue distribution and expression levels

    • Variations in regulatory kinase activities

    • Divergent metabolic regulation across species

  • Methodological translation:

    • Validation of antibody cross-reactivity with human samples

    • Optimization of fixation protocols for human specimens

    • Accounting for post-mortem changes in phosphorylation

    • Development of clinical-grade detection methods

  • Physiological context differences:

    • Variations in basal phosphorylation states

    • Different stress response mechanisms

    • Longer timeframes for human disease progression

    • Comorbidities and polypharmacy in human patients

  • Disease model relevance:

    • Fidelity of animal models to human disease mechanisms

    • Different rates of disease progression

    • Variation in drug metabolism and pharmacokinetics

    • Ethical limitations in human experimental interventions

  • Translational strategy development:

    • Employ humanized animal models where appropriate

    • Validate findings in human primary cell cultures

    • Use patient-derived xenografts for cancer studies

    • Develop surrogate biomarkers for clinical studies

While BAD sequence is relatively conserved between species, particularly around the S155 phosphorylation site, regulatory mechanisms and disease contexts can vary significantly, necessitating careful validation of findings when translating from animal models to human applications.

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.