Phospho-HSPB1 (S78) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Mechanism of Action and Biological Relevance

Phosphorylation of HSPB1 at serine 78 (S78) regulates its chaperone activity, subcellular localization, and interaction with stress-responsive pathways. This modification is mediated by kinases such as MAPKAP kinase-2 and is critical for:

  • Stress Adaptation: Enhancing cytoskeletal stability and protecting cells against oxidative stress, heat shock, and chemotherapy-induced damage .

  • Protein Quality Control: Maintaining denatured proteins in a folding-competent state and preventing aggregation .

  • Disease Pathways: Dysregulation of S78 phosphorylation is implicated in cancer progression, neurodegenerative disorders, and neuropathies like Charcot-Marie-Tooth disease .

Stress Response and Cellular Signaling

The antibody has been pivotal in elucidating HSPB1’s role in stress adaptation:

  • Heat Shock and Oxidative Stress: Phosphorylation at S78 increases under stress, enhancing HSPB1’s ability to stabilize actin filaments and protect against cytoskeletal disruption .

  • Cancer Biology: Elevated S78 phosphorylation correlates with cancer cell survival, metastasis, and resistance to apoptosis. Studies using this antibody identified HSPB1 as a therapeutic target in breast, cervical, and lung cancers .

Neurodegenerative and Neuropathic Disorders

Mutations in HSPB1 (e.g., R127W, S135F) disrupt oligomerization and phosphorylation dynamics, leading to Charcot-Marie-Tooth disease. This antibody has enabled researchers to:

  • Map Phosphorylation-Dependent Interactions: Demonstrate how S78 phosphorylation modulates HSPB1’s binding to neurofilaments and axonal transport proteins .

  • Assess Disease-Associated Mutants: Compare wild-type and mutant HSPB1’s phosphorylation patterns to understand pathogenic mechanisms .

Clinical and Therapeutic Implications

  • Biomarker Potential: Phospho-HSPB1 (S78) levels correlate with tumor aggressiveness and treatment resistance, positioning it as a prognostic marker in oncology .

  • Therapeutic Targeting: Inhibiting S78 phosphorylation may disrupt stress adaptation pathways in cancer cells, offering a novel strategy for combination therapies .

Technical Considerations

  • Validation: Ensure proper antigen retrieval (e.g., citrate buffer pH 6.0) for IHC and use blocking peptides to confirm specificity .

  • Storage: Maintain at -20°C to preserve activity, avoiding repeated freeze-thaw cycles .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

This phospho-HSPB1 (S78) recombinant monoclonal antibody is manufactured through a multi-step process. It begins with the retrieval of genes encoding the HSPB1 antibody from rabbits previously immunized with a synthetic peptide derived from the human HSPB1 protein phosphorylated at S78. These genes are then integrated into specialized expression vectors, which are subsequently introduced into host suspension cells. These cells are then cultivated to induce the production and secretion of the antibody. Following this growth phase, the phospho-HSPB1 (S78) recombinant monoclonal antibody is purified using affinity chromatography techniques, separating it from the cell culture supernatant. Finally, the antibody's effectiveness is rigorously assessed through a series of assays, including ELISA, Western blotting, and immunohistochemistry, confirming its ability to interact specifically with the human HSPB1 protein phosphorylated at S78.

Phosphorylation of HSPB1 at S78 is a critical regulatory mechanism that enables cells to respond to stress, maintain protein quality control, and promote cell survival. Disruption of this phosphorylation event can lead to significant implications in various diseases, including cancer and neurodegenerative disorders.

Form
Liquid
Lead Time
We can typically dispatch the products within 1-3 working days after receiving your order. The delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
Heat shock 27kDa protein antibody; 28 kDa heat shock protein antibody; CMT2F antibody; DKFZp586P1322 antibody; epididymis secretory protein Li 102 antibody; Estrogen regulated 24 kDa protein antibody; Estrogen-regulated 24 kDa protein antibody; Heat shock 25kDa protein 1 antibody; Heat shock 27 kDa protein antibody; Heat shock 27kD protein 1 antibody; Heat shock 27kDa protein 1 antibody; Heat shock 28kDa protein 1 antibody; Heat Shock Protein 27 antibody; Heat shock protein beta 1 antibody; Heat shock protein beta-1 antibody; heat shock protein family B (small) member 1 antibody; HEL-S-102 antibody; HMN2B antibody; HS.76067 antibody; Hsp 25 antibody; HSP 27 antibody; Hsp 28 antibody; Hsp B1 antibody; Hsp25 antibody; HSP27 antibody; Hsp28 antibody; HspB1 antibody; HSPB1_HUMAN antibody; SRP27 antibody; Stress responsive protein 27 antibody; Stress-responsive protein 27 antibody
Target Names
Uniprot No.

Target Background

Function

HSPB1, also known as Hsp27, is a small heat shock protein that functions as a molecular chaperone. It plays a crucial role in maintaining denatured proteins in a folding-competent state, contributing to stress resistance and actin organization. Through its chaperone activity, HSPB1 may regulate various biological processes, including the phosphorylation and axonal transport of neurofilament proteins.

Gene References Into Functions
  1. Research suggests that both the small heat shock protein HspB1/Hsp27 and the constitutive chaperone Hsc70/HspA8 interact with tau to prevent tau-fibril/amyloid formation. HspB1 delays tau-fibril formation by weakly interacting with early species in the aggregation process. (HspB1 = heat shock protein family B small member 1; Hsc70 = heat shock protein family A Hsp70) PMID: 29298892
  2. HSP27 is a partner of JAK2-STAT5 and a potential therapeutic target in myelofibrosis. PMID: 29650953
  3. Results demonstrate that HSP27 expression is significantly increased in metastatic prostate cancer and serves as a strong marker predicting survival, along with ALDH6A1 and prohibitin. PMID: 30396985
  4. High HSP27 expression is associated with breast cancer. PMID: 30106436
  5. Studies suggest that overexpression of peroxiredoxin-2, annexin A2, and heat shock protein beta-1 was correlated with tumor invasion, metastasis, and poor prognosis, indicating their potential as diagnostic and therapeutic biomarkers. PMID: 29332450
  6. ETAS down-regulated HSP27 and pHSP27 (serine 78) in KLM1-R cells, while HSP70 and GRP78 levels remained unchanged. This research suggests the potential therapeutic benefit of ETAS in enhancing anticancer effects when combined with gemcitabine for pancreatic cancer patients. PMID: 29936456
  7. Silencing of HSP27 increases curcumin resistance by reducing apoptosis and reactive oxidative stress production in colon cancer. PMID: 30056019
  8. The expression of type I and III collagen in the TGF-beta1-induced transition of A549 cells to myofibroblasts was significantly downregulated by transfection of A549 cells with HSP27 plasmid Dinterfering sequence and an optimal ratio of Lipofectamine(R) 2000 and HSP27 plasmid. PMID: 29568951
  9. It was suggested that HSP27 expression in EUS-FNA samples could be useful for predicting GEM sensitivity. PMID: 29695571
  10. High hsp27 expression is associated with leukemia. PMID: 29328466
  11. p-Hsp27 is a novel biomarker of glioma and may have important clinical value for further classification of patients with wild-type IDH1 and normal ATRX expression and for evaluating prognosis. PMID: 29550762
  12. Dimer-monomer equilibrium of human HSP27 is influenced by the in-cell macromolecular crowding environment and is controlled by fatty acids and heat. PMID: 29635040
  13. HSP27 expression promotes vascular smooth muscle cell viability, suppresses cell apoptosis, and confers protection against oxidative stress in Thoracic aortic dissection. PMID: 29209372
  14. Hsp27 and P38MAPK could be used as prognostic factors in Esophageal squamous cell carcinoma. PMID: 29099815
  15. Addition of apatorsen to chemotherapy does not improve outcomes in unselected patients with metastatic pancreatic cancer in the first-line setting. However, a trend toward prolonged PFS and OS in patients with high baseline serum Hsp27 suggests that this therapy may warrant further evaluation in this subgroup. PMID: 28935773
  16. High HSP27 expression is associated with non-small cell lung cancer metastasis. PMID: 28656293
  17. Knockdown of HSP27 blocked TGF-beta-induced cisplatin resistance. PMID: 28848138
  18. The expression of HSP27 was approximately 2-fold higher in apical periodontitis. Furthermore, an increased expression of HSP27 was detected in epithelial cells. PMID: 28673495
  19. Findings indicate the heat-shock protein 27 (Hsp27) pathway as a therapeutic target for the management of conditions featuring dysregulated interleukin 1 beta (IL-1beta) production. PMID: 27976724
  20. Increased protein level of HSP27 through SUMO2/3-mediated SUMOylation plays crucial roles in the progression of primary hepatocellular carcinoma. PMID: 28665748
  21. When correlations of the markers with the response to neoadjuvant chemotherapy were examined, only high pre-chemotherapy levels of cytoplasmic HSPB1/p correlated with a poor clinical and pathological response to neoadjuvant cisplatin chemotherapy (p = 0.056) suggesting that this marker could be useful for further study in a larger number of cases. PMID: 28608263
  22. Cetuximab, in combination with various concentrations of AG490, an inhibitor of JAK2, STAT3 and HSP27 protein levels, except in the KRAS G12V mutant line, SW620...cetuximab may promote SN38 sensitivity via suppression of HSP27, through blocking the JAK/STAT signaling pathway, and shows synergistic effects when combined with SN38 in wild-type RAS CRC cells. PMID: 28656305
  23. Research has concluded that HSP27-silenced placenta-derived multipotent cells differentiated into neurons possessing the characteristics of functional glutamatergic neurons. PMID: 27444754
  24. Data show that Hsp27 increases the degradation rate of ubiquitinated MST1 and therefore interrupts the Hippo pathway kinase cascade. Consequently, YAP and TAZ are less phosphorylated, becoming free to translocate into the nucleus, promoting a malignant phenotype. These findings highlight the crucial role of Hsp27 in regulating multiple signaling pathways that contribute to tumor aggressiveness. PMID: 27555231
  25. Upregulation of Hsp27 is a common phenomenon shared between pregnancies in patients with preterm prelabor rupture of membranes and spontaneous preterm labor with intact membranes. PMID: 28497897
  26. HSP27 is an independent predictor of prognosis in chronic HF. PMID: 28844461
  27. Hsp27 may up-regulate the expression of ABCA1 and promotes cholesterol efflux through activation of the PI3K/PKCzeta/Sp1 signal pathway in THP-1 macrophage-derived foam cells. PMID: 28610841
  28. This study reports solution-state nuclear magnetic resonance spectroscopy investigations of the conformation and dynamics of the disordered and flexible C-terminal region of human HSP27. These data indicate a potential role for cis-trans proline isomerization in regulating oligomerization. PMID: 28547731
  29. Overexpression of both HSPB5 and Hsp27 significantly reduced the intracellular aggregation of alpha-synuclein. PMID: 28337642
  30. HspB1 structural organization displays dynamic and complex rearrangements in response to changes in the cellular environment or when cell physiology is modified. [review] PMID: 28144778
  31. Glutathione-S-transferase - HspB1 fusion protein prevents more aggregation of malate dehydrogenase compared to glutathione-S-transferase -HspB5 fusion protein and wild type HspB1. PMID: 28130664
  32. Data suggest that oncogene-addicted cells require the small heat-shock protein of 27 kDa (HSP27) for survival and that HSP27 might interfere with the effectiveness of targeted agents. PMID: 28182330
  33. Research has identified six cytostatic drugs that inhibit heat shock 27 kDa protein (HSP27) and tackle drug resistance using a computational drug repositioning approach. PMID: 27626687
  34. This condition is distinct from the C-M-T phenotype observed in hereditary neuropathies caused by mutations in the HSPB1 gene. PMID: 27816334
  35. Studies have shown that knock down of HSPB1 further increased the proportion of apoptotic cells in hyperthermic treated melanoma cells compared to either single agent alone, and both agents led to cell cycle arrest at G0/G1 or G2/M phases. PMID: 27626679
  36. Data suggest that altered dimerization of HSP27 represents a promising strategy for anticancer therapy in HSP27-overexpressing cancer cells. PMID: 27449291
  37. Apatorsen (2'-methoxyethyl-modified antisense oligonucleotide that inhibits Hsp27 expression) was tolerated at the highest dose evaluated (1000 mg). Single-agent activity was suggested by changes in tumor markers, circulating tumor cells, and stable measurable disease. PMID: 27022067
  38. This research demonstrates the central role of Hsp27 in RACK1 pseudosubstrate or LPS-induced cell activation of primary leukocytes. PMID: 27178349
  39. High p-Hsp27 expression is associated with cisplatin resistance in lung cancer. PMID: 26872057
  40. Low p-Hsp27 expression is associated with pancreatic cancer. PMID: 26895107
  41. The up-regulation of Hsp27 by E2 is mediated by ERalpha/Sp1. PMID: 27179757
  42. A study reports a novel interaction between mutant HSPB1-P182L and the RNA binding protein PCBP1, leading to a reduction in its translational repression activity. Identifying PCBP1 mRNA targets revealed a marked prevalence for an RNA recognition motif, preferably seen in their 5' and 3'UTRs. Findings further support a role for mutant HSPB1 in neurodegenerative diseases. PMID: 28077174
  43. Hsp 70 and Hsp 27 were expressed in middle ear effusions. PMID: 28964286
  44. High Hsp27 expression is associated with Thyroid Tumors. PMID: 26970173
  45. Bradykinin stimulates myofibroblast migration through protein kinase D-mediated activation of COX-2 and Hsp27. PMID: 28032559
  46. Data suggest that HSPB6 forms hetero-oligomers with HSPB1 under the following rules: (1) highly conserved motif RLFDQXFG is necessary for subunit exchange among oligomers, (2) a site about 20 residues downstream of this motif determines the size of resultant hetero-oligomers, and (3) a region in the N-terminal domain that is unique to HSPB6 dictates preferential formation of heterodimers. (HSP = heat shock protein) PMID: 28487364
  47. TNF-alpha stimulates colonic myofibroblast migration through P38 MAPK-mediated activation of COX-2 and Hsp27. PMID: 27451881
  48. The potency of the sHsps to suppress aggregation of model substrates is correlated with their ability to form stable substrate complexes; especially HspB1 and HspB5, but also B3, bind tightly to a variety of proteins. PMID: 27909051
  49. This work examines the molecular mechanism by which two canonical sHsps, alphaB-crystallin (alphaB-c) and Hsp27, interact with aggregation-prone alpha-syn to prevent its aggregation in vitro. Both sHsps are very effective inhibitors of alpha-syn aggregation. PMID: 27587396
  50. This study highlights a novel relation between serum pHSP27 and BMD in Indian women; however, these findings need to be confirmed in larger studies. PMID: 27241641

Show More

Hide All

Database Links

HGNC: 5246

OMIM: 602195

KEGG: hsa:3315

STRING: 9606.ENSP00000248553

UniGene: Hs.520973

Involvement In Disease
Charcot-Marie-Tooth disease 2F (CMT2F); Neuronopathy, distal hereditary motor, 2B (HMN2B)
Protein Families
Small heat shock protein (HSP20) family
Subcellular Location
Cytoplasm. Nucleus. Cytoplasm, cytoskeleton, spindle.
Tissue Specificity
Detected in all tissues tested: skeletal muscle, heart, aorta, large intestine, small intestine, stomach, esophagus, bladder, adrenal gland, thyroid, pancreas, testis, adipose tissue, kidney, liver, spleen, cerebral cortex, blood serum and cerebrospinal f

Q&A

Structural and Functional Characteristics of HSPB1

Q: What is HSPB1 and what is its fundamental role in cellular physiology?

A: HSPB1 (Heat Shock Protein Beta-1), also known as HSP27, is an ATP-independent, small-sized (approximately 27 kDalton) chaperone molecule belonging to the small heat shock protein (HSP20) family. It exists at basal levels in cells and tissues, typically organized as large oligomers reaching up to 1,000 kD in size. HSPB1 plays crucial roles in cellular stress response mechanisms, including protection against heat shock, hypoxia, oxidative stress, infections, and ultraviolet radiation. The protein functions primarily as a molecular chaperone that prevents protein aggregation, assists in protein folding, and contributes to cellular homeostasis. In non-stressed conditions, HSPB1 maintains cytoskeletal integrity and regulates apoptotic pathways. Under stress conditions, its expression increases significantly to protect cellular proteins from damage and maintain cellular viability .

Q: How does phosphorylation alter the structure and function of HSPB1?

A: Phosphorylation represents a critical regulatory mechanism for HSPB1 function. In its unphosphorylated state, HSPB1 exists predominantly as large oligomeric complexes. Following phosphorylation at specific serine residues (Ser-15, Ser-78, Ser-82, Thr-143), HSPB1 undergoes a significant structural reorganization, dissociating into smaller oligomers, primarily dimers and tetramers. This structural transition fundamentally alters the protein's function and interaction capabilities. The phosphorylated smaller units can interact with different protein partners than the unphosphorylated larger oligomers, effectively redirecting HSPB1's cellular activities. This simple change in phosphorylation state regulates many of the canonical functions of HSPB1, including its chaperone activity, cytoskeletal organization, and anti-apoptotic functions. Phosphorylation also affects HSPB1's cellular localization, with phosphorylated forms showing distinct intracellular distribution patterns compared to the unphosphorylated protein .

Significance of HSPB1 S78 Phosphorylation

Q: Why is the phosphorylation of HSPB1 at Serine 78 specifically important to research?

A: Phosphorylation of HSPB1 at Serine 78 represents a critical regulatory mechanism with significant research implications. This specific phosphorylation event triggers important structural changes that alter HSPB1's functional properties. When phosphorylated at S78, HSPB1 reorganizes from large oligomers into smaller functional units, enabling distinct protein-protein interactions and modified cellular activities. This phosphorylation site is particularly important because it allows cells to respond appropriately to stress, maintain protein quality control, and promote cell survival under challenging conditions .

The S78 phosphorylation state serves as a valuable biomarker in various pathological contexts. Dysregulation of this phosphorylation event has significant implications in numerous diseases, including cancer and neurodegenerative disorders. In cancer research, increased HSPB1 expression and phosphorylation have been linked to tumor progression, metastatic potential, and treatment resistance, making it a promising therapeutic target. The ability to specifically detect and quantify S78 phosphorylation provides researchers with a powerful tool to investigate these disease-related pathways and potentially develop targeted interventions .

Q: What signaling pathways regulate HSPB1 phosphorylation at S78?

A: The phosphorylation of HSPB1 at Serine 78 is primarily regulated through stress-activated protein kinase cascades. The p38 MAPK pathway plays a central role in this process, where environmental stressors activate p38 MAPK, which subsequently phosphorylates and activates MAPKAPK2 (MK2) and MAPKAPK3 (MK3). These kinases directly phosphorylate HSPB1 at S78, as demonstrated in experimental systems using anisomycin (a p38 MAPK activator) and PF-3644022 (a MAPKAPK2 inhibitor). When cells are treated with anisomycin, researchers observe increased phosphorylation of HSPB1 at S78, while co-treatment with PF-3644022 causes a dose-dependent decrease in this phosphorylation without affecting total HSPB1 levels .

Additionally, other kinases including p70S6K, Protein Kinase G (PKG), and Protein Kinase C (PKC) have been implicated in HSPB1 phosphorylation under specific cellular conditions, suggesting multiple regulatory inputs converge on this critical modification. This complex regulation allows for fine-tuning of HSPB1 function in response to diverse cellular stresses and signaling events .

Applications of Phospho-HSPB1 (S78) Antibodies

Q: What are the primary research applications for Phospho-HSPB1 (S78) antibodies?

A: Phospho-HSPB1 (S78) antibodies serve as versatile tools across multiple research applications:

  • Western Blot (WB): These antibodies are optimized for Western blot applications with recommended dilutions typically ranging from 1:500 to 1:5000, making them ideal for quantitative analysis of phosphorylation states across different experimental conditions. Western blot allows researchers to confirm the specificity of the antibody by verifying the molecular weight of the detected protein (observed at approximately 28kDa despite a calculated MW of 23kDa) .

  • Immunohistochemistry (IHC): With dilutions ranging from 1:50 to 1:200, these antibodies enable visualization of phosphorylated HSPB1 in tissue sections, providing insights into its spatial distribution and expression patterns in different physiological and pathological contexts .

  • Immunofluorescence/Immunocytochemistry (IF/ICC): Using dilutions of approximately 1:100 to 1:200, researchers can examine the subcellular localization of phosphorylated HSPB1, which is typically found in the cytoplasm, nucleus, cytoskeleton, and spindle structures .

  • ELISA: These antibodies are compatible with ELISA techniques for quantitative measurement of phosphorylated HSPB1 levels in cell and tissue lysates .

  • HTRF (Homogeneous Time-Resolved Fluorescence) Assays: Modern phospho-specific antibodies are incorporated into HTRF technology, offering a streamlined, plate-based approach without requiring gels, electrophoresis, or transfers. This method demonstrates significantly higher sensitivity (up to 32 times more sensitive than Western blot) and requires fewer cells (120 cells/well compared to 3,750 cells needed for Western blot) .

These applications collectively allow researchers to study signaling pathways, cellular stress responses, and potential therapeutic targets related to HSPB1 phosphorylation across various experimental systems.

Antibody Selection and Validation

Q: What criteria should I consider when selecting between monoclonal and polyclonal Phospho-HSPB1 (S78) antibodies?

A: When selecting between monoclonal and polyclonal Phospho-HSPB1 (S78) antibodies, researchers should consider several important factors:

Monoclonal Antibodies (e.g., Recombinant Monoclonal):

  • Higher specificity for the phosphorylated S78 epitope, reducing cross-reactivity with other phosphorylation sites

  • Greater consistency between batches, enabling more reproducible results across long-term studies

  • Production process typically involves retrieving genes encoding the HSPB1 antibody from rabbits exposed to synthesized phosphopeptides, followed by integration into expression vectors, introduction into host suspension cells, and purification via affinity chromatography

  • Ideal for quantitative applications where precision and reproducibility are essential

  • Generally preferred for clinical research applications due to their consistent performance characteristics

Polyclonal Antibodies:

  • Recognize multiple epitopes around the phosphorylated S78 site, potentially providing stronger signals

  • Typically produced by immunizing rabbits with synthetic phosphorylated peptides around S78 of human HSPB1

  • May offer greater sensitivity for detecting low abundance targets as they can bind to multiple epitopes on the same molecule

  • Potentially more tolerant of minor protein denaturation or conformational changes

  • Useful for initial exploratory research where the signal strength may be prioritized over absolute specificity

For validation, both antibody types should be tested against positive controls (e.g., HeLa cells treated with anisomycin to induce HSPB1 phosphorylation) and negative controls (e.g., HAP1 knock-out HSP27 cell lines). The selection ultimately depends on the specific research application, with monoclonal antibodies generally preferred for precise quantification and polyclonal antibodies often chosen for detection of low-abundance targets or preliminary investigations .

Experimental Design Considerations

Q: How should I design experiments to investigate the dynamic phosphorylation patterns of HSPB1 at S78 compared to other phosphorylation sites?

A: Designing experiments to investigate dynamic phosphorylation patterns of HSPB1 requires a multi-faceted approach:

  • Temporal Analysis: Implement time-course experiments (5, 15, 30, 60 minutes) after stimulus application to capture the kinetics of phosphorylation across different sites (S15, S78, S82, T143). This approach reveals whether S78 phosphorylation precedes or follows other modifications, providing insight into the sequential regulation of HSPB1 function.

  • Parallel Site Monitoring: Utilize site-specific antibodies against all major phosphorylation sites simultaneously in parallel samples. This comparative analysis requires careful antibody validation to ensure specificity for each phospho-site without cross-reactivity.

  • Stimulus Specificity: Compare diverse cellular stressors (oxidative stress, heat shock, inflammatory cytokines, UV radiation) to determine if S78 phosphorylation responds differentially compared to other sites. For example, anisomycin effectively induces S78 phosphorylation through p38 MAPK pathway activation .

  • Kinase Inhibitor Panel: Apply specific inhibitors targeting relevant kinases (p38 MAPK inhibitors, MAPKAPK2 inhibitors like PF-3644022, PKC inhibitors) to dissect the regulatory pathways controlling each phosphorylation site. This reveals whether S78 phosphorylation operates through distinct or overlapping signaling networks compared to other sites .

  • Phosphomimetic Mutations: Employ HSPB1 constructs with phosphomimetic mutations (S→D/E) or phospho-null mutations (S→A) at individual or combined sites to analyze functional consequences of specific phosphorylation patterns.

  • Mass Spectrometry Validation: Confirm antibody-based results with quantitative phosphoproteomic analysis to provide unbiased measurement of all phosphorylation sites simultaneously, allowing precise stoichiometric calculations of modification levels.

This integrated approach enables comprehensive characterization of S78 phosphorylation dynamics in relation to other HSPB1 modifications, revealing their cooperative or antagonistic relationships in different cellular contexts.

Disease Model Applications

Q: How can I effectively study HSPB1 S78 phosphorylation in cancer research models?

A: Studying HSPB1 S78 phosphorylation in cancer research requires strategic approaches across multiple experimental systems:

  • Cell Line Selection and Characterization: Begin by screening a panel of cancer cell lines (A549, HeLa, and tissue-specific lines) for baseline HSPB1 expression and S78 phosphorylation using HTRF or Western blot techniques. Establish quantitative baselines using the HTRF Phospho-HSP27 (Ser78) detection kit, which provides superior sensitivity compared to traditional Western blot (detecting signal from as few as 120 cells/well) .

  • Modulation of HSPB1 Phosphorylation: Employ pharmacological interventions targeting the p38 MAPK pathway, such as anisomycin for activation and PF-3644022 (MAPKAPK2 inhibitor) for inhibition. Monitor dose-dependent effects on S78 phosphorylation while simultaneously assessing total HSPB1 levels and cell viability to distinguish between effects on phosphorylation versus protein expression .

  • Genetic Manipulation Strategies: Implement CRISPR/Cas9 knockout systems or RNAi approaches to modulate HSPB1 expression. Compare results with HAP1 knock-out HSP27 cell lines as validated controls. For mechanistic studies, introduce phospho-null (S78A) or phosphomimetic (S78D/E) HSPB1 mutants to examine functional consequences .

  • Correlation with Cancer Phenotypes: Analyze how S78 phosphorylation status correlates with cancer hallmarks including proliferation, invasion, metastatic potential, and treatment resistance. Design experiments to measure these parameters following modulation of S78 phosphorylation.

  • Patient-Derived Models: Extend findings to patient-derived xenografts or organoids, which better recapitulate tumor heterogeneity. Measure S78 phosphorylation in these models and correlate with clinical parameters including treatment response and patient outcomes.

  • Therapeutic Implications: Evaluate how existing cancer therapeutics affect HSPB1 S78 phosphorylation, and conversely, how modulating S78 phosphorylation impacts therapeutic efficacy. This bidirectional analysis can identify potential combination strategies targeting HSPB1 phosphorylation to overcome treatment resistance.

This comprehensive approach generates mechanistically informative data on HSPB1 S78 phosphorylation in cancer that may translate to novel therapeutic strategies targeting this modification.

Signaling Pathway Analysis

Q: What techniques can I use to elucidate the complete signaling networks connecting stress stimuli to HSPB1 S78 phosphorylation?

A: Elucidating the complete signaling networks regulating HSPB1 S78 phosphorylation requires an integrated approach combining targeted interventions with systems-level analysis:

  • Kinase Activity Profiling: Implement a focused kinase inhibitor screen using compounds targeting known regulators (p38 MAPK, MAPKAPK2/3, PKG, PKC) and potential novel kinases. The inhibitor PF-3644022 (MAPKAPK2-specific) produces a dose-dependent decrease in S78 phosphorylation following anisomycin treatment, confirming this pathway's involvement. Quantify phosphorylation using HTRF assays, which offer superior sensitivity compared to Western blotting .

  • Phosphoproteomic Analysis: Employ mass spectrometry-based phosphoproteomics to simultaneously monitor hundreds of phosphorylation events following stimulus application. This approach identifies both known and unexpected signaling nodes activated concurrently with HSPB1 S78 phosphorylation, revealing potential new regulatory connections.

  • Genetic Perturbation Strategies: Apply CRISPR-based screens or RNAi libraries targeting signaling components to identify genes whose disruption affects S78 phosphorylation. Compare results with established knockout models like HAP1 HSP27 KO cell lines as standards for complete phosphorylation absence .

  • Temporal Signaling Dynamics: Implement high-resolution time-course experiments measuring phosphorylation of HSPB1 and upstream kinases (p38 MAPK, MAPKAPK2) following stress application. The timing of phosphorylation events reveals the sequence of activation within the signaling cascade.

  • Pathway Crosstalk Analysis: Investigate interactions between p38 MAPK and other stress-response pathways by simultaneously monitoring multiple signaling outputs. This reveals how HSPB1 S78 phosphorylation integrates signals from various cellular stress sensors.

  • Proximity Labeling Approaches: Apply BioID or APEX2-based proximity labeling with HSPB1 as the bait to identify proteins physically associated with HSPB1 during stress responses. This approach reveals both upstream kinases and downstream effectors in their native cellular context.

This multi-dimensional approach generates a comprehensive map of signaling networks controlling HSPB1 S78 phosphorylation, providing insights into both canonical and non-canonical regulatory mechanisms.

Sample Preparation Protocols

Q: What are the optimal sample preparation methods for preserving HSPB1 phosphorylation status in various experimental systems?

A: Preserving HSPB1 phosphorylation status during sample preparation is critical for accurate analysis and requires careful attention to multiple factors:

  • Cell Harvesting and Lysis:

    • Utilize rapid harvesting techniques to minimize dephosphorylation during processing.

    • Lyse cells directly in the culture plate using supplemented Lysis Buffer #1 (as specified in HTRF protocols) for 30 minutes at room temperature under gentle shaking .

    • Maintain consistent cell density across experimental conditions (approximately 12,500 cells/well in 96-well format has been validated for both A549 and HeLa cells) .

    • Include phosphatase inhibitors (sodium fluoride, sodium orthovanadate, β-glycerophosphate) in all buffers to prevent dephosphorylation events.

  • Buffer Composition:

    • Ensure lysis buffers contain detergents suitable for releasing both cytoplasmic and nuclear proteins.

    • Maintain physiological pH (7.2-7.4) during lysis to prevent artificial alterations in phosphorylation.

    • Add protease inhibitors to prevent degradation of HSPB1 protein.

  • Temperature Control:

    • Process samples consistently at either 4°C (to minimize enzymatic activity) or room temperature following validated protocols.

    • Avoid freeze-thaw cycles which can affect phosphorylation status; aliquot lysates immediately after preparation.

  • Sample Storage:

    • For short-term storage (1-3 days), maintain samples at -20°C.

    • For long-term preservation, store at -80°C with glycerol addition (10-15%) to prevent freeze-damage.

    • When using stored samples, thaw rapidly at room temperature and process immediately.

  • Sample Dilution Strategies:

    • Optimize dilution factors based on cell type and expected phosphorylation levels (e.g., 1/2 and 1/8 dilutions for Phospho-HSP27 S78, 1/4 and 1/8 for Total HSP27 in A549 and HeLa samples) .

    • Maintain consistent protein concentration across comparative samples to ensure quantitative accuracy.

These methodological considerations ensure that the phosphorylation status observed experimentally accurately reflects the biological state of HSPB1 in the original sample, avoiding artifacts introduced during processing.

Antibody Optimization Strategies

Q: How should I optimize Phospho-HSPB1 (S78) antibody conditions for different experimental techniques?

A: Optimizing Phospho-HSPB1 (S78) antibody conditions requires technique-specific approaches:

  • Western Blot Optimization:

    • Begin with the manufacturer-recommended dilution range (1:500-1:5000) and systematically test multiple dilutions to determine optimal signal-to-noise ratio .

    • Implement gradient exposure times to identify the linear detection range for quantitative analysis.

    • Compare blocking agents (BSA vs. non-fat milk) as milk proteins contain phosphatases that may interfere with phospho-epitope detection.

    • Validate specificity using positive controls (anisomycin-treated cells) and negative controls (HAP1 HSP27 knockout cells) .

    • Verify the observed molecular weight matches expectations (approximately 28kDa despite a calculated MW of 23kDa) .

  • Immunohistochemistry (IHC) Optimization:

    • Start within the recommended dilution range (1:50-1:200) and systematically refine based on signal intensity and specificity .

    • Test multiple antigen retrieval methods (heat-induced vs. enzymatic) to maximize epitope accessibility.

    • Optimize incubation conditions (time, temperature) to enhance specific binding while minimizing background.

    • Compare detection systems (chromogenic vs. fluorescent) for optimal visualization of the phospho-specific signal.

  • Immunofluorescence/Immunocytochemistry (IF/ICC) Optimization:

    • Begin with recommended dilutions (1:100-1:200) and adjust based on signal intensity .

    • Test fixation methods (paraformaldehyde, methanol, acetone) to determine which best preserves the phospho-epitope.

    • Implement antigen retrieval techniques if necessary to enhance epitope accessibility.

    • Include counterstains to verify the expected subcellular localization (cytoplasm, nucleus, cytoskeleton, spindle) .

  • HTRF Assay Optimization:

    • Determine optimal cell lysate dilutions based on cell type (different dilutions required for A549 vs. HeLa cells) .

    • Establish appropriate incubation times (4 hours to overnight) to maximize signal development .

    • Validate results across multiple readers (Envision Nexus, standard HTRF readers) to ensure instrumentation compatibility.

  • Cross-Technique Validation:

    • Compare results across multiple detection methods to confirm consistent findings.

    • Implement quantitative analysis to establish the relative sensitivity of different techniques (HTRF being approximately 32 times more sensitive than Western blot) .

This systematic optimization approach ensures robust and reproducible detection of HSPB1 S78 phosphorylation across diverse experimental platforms.

Control Selection and Validation

Q: What are the essential controls for experiments investigating HSPB1 S78 phosphorylation?

A: Implementing comprehensive controls is critical for establishing the validity and interpretability of HSPB1 S78 phosphorylation experiments:

Implementation of these comprehensive controls ensures experimental rigor and enables confident interpretation of HSPB1 S78 phosphorylation data across diverse experimental contexts.

HTRF vs Western Blot Analysis

Q: What are the comparative advantages of HTRF over Western blot for detecting HSPB1 S78 phosphorylation?

A: HTRF (Homogeneous Time-Resolved Fluorescence) technology offers several significant advantages over traditional Western blot for detecting HSPB1 S78 phosphorylation:

  • Sensitivity:

    • HTRF demonstrates dramatically higher sensitivity, requiring as few as 120 cells/well to detect significant phospho-HSPB1 (S78) signal, compared to Western blot which requires at least 3,750 cells for minimal detection. This represents approximately 32-fold higher sensitivity with HTRF technology .

    • This enhanced sensitivity enables experiments with limited biological material or rare cell populations.

  • Workflow Efficiency:

    • HTRF is a plate-based "no-wash" assay that eliminates labor-intensive steps required for Western blot (gel electrophoresis, transfer, blocking, multiple washing steps).

    • The streamlined HTRF workflow can be completed in a single plate used for culturing, stimulation, and detection, significantly reducing hands-on time .

    • Results can be obtained within 4 hours of incubation, compared to Western blot which typically requires 1-2 days for completion .

  • Quantitative Accuracy:

    • HTRF provides precise quantification across a wider dynamic range compared to Western blot, which often suffers from saturation at higher expression levels.

    • The ratiometric measurement (665nm/620nm) in HTRF corrects for well-to-well variations, providing more consistent results compared to Western blot densitometry.

  • Reproducibility:

    • HTRF demonstrates lower coefficient of variation between replicates and across experiments compared to Western blot, which is subject to transfer efficiency variability and other technical inconsistencies.

    • The homogeneous nature of HTRF reactions minimizes technical variations introduced by multiple handling steps.

  • Multiplexing Capacity:

    • HTRF can be multiplexed with other assays in the same experimental setup, such as simultaneously measuring cell viability (ATPlite) alongside phosphorylation status .

    • This enables direct correlation between phosphorylation events and functional outcomes within the same cell population.

  • High-Throughput Compatibility:

    • HTRF is readily adaptable to high-throughput screening formats, enabling testing of numerous conditions, compounds, or genetic perturbations in parallel.

    • The 384-well format is standardized for HTRF applications, supporting miniaturization and resource optimization .

This comprehensive comparison demonstrates that while Western blot remains valuable for initial validation and molecular weight confirmation, HTRF offers superior performance for quantitative analysis of HSPB1 S78 phosphorylation, particularly in applications requiring sensitivity, throughput, or precise quantification.

Multi-parameter Detection Methods

Q: How can I simultaneously measure HSPB1 S78 phosphorylation along with other signaling pathway components?

A: Implementing multi-parameter detection methods enables comprehensive analysis of HSPB1 S78 phosphorylation within its broader signaling context:

  • Multiplexed HTRF Approaches:

    • Design experimental workflows to measure phospho-HSPB1 (S78), total HSPB1, and upstream kinases (phospho-p38 MAPK Thr180/Tyr182) from the same cell population using sample splitting at appropriate dilutions (neat and 1/2 for phospho-p38 MAPK, 1/2 and 1/8 for phospho-HSPB1, 1/4 and 1/8 for total HSPB1) .

    • Incorporate cell viability assessment (ATPlite) in parallel to distinguish signaling changes from cytotoxic effects .

    • This approach enables direct correlation between p38 MAPK activation, HSPB1 phosphorylation, and cellular phenotypes.

  • Multi-Color Flow Cytometry/Mass Cytometry:

    • Develop antibody panels using differently labeled phospho-specific antibodies (phospho-HSPB1 S78, phospho-p38 MAPK, phospho-MK2) to analyze signaling at the single-cell level.

    • This approach reveals cell-to-cell heterogeneity in signaling responses and identifies distinct cellular subpopulations based on their phosphorylation profiles.

    • Combines with surface markers to correlate phosphorylation status with cell type or activation state.

  • Multiplex Immunoassay Systems:

    • Employ bead-based multiplex immunoassay platforms to simultaneously quantify multiple phosphoproteins from limited sample volumes.

    • This methodology allows comprehensive pathway analysis, measuring phosphorylation of HSPB1 at multiple sites (S15, S78, S82) together with upstream and downstream signaling proteins.

  • Automated Western Blot Systems:

    • Utilize capillary-based automated Western systems for sequential reprobing of the same samples with multiple phospho-specific antibodies without membrane stripping.

    • This approach provides molecular weight confirmation while enabling comparative quantification across multiple targets.

  • High-Content Imaging:

    • Implement immunofluorescence protocols with multiplexed antibody detection to visualize phospho-HSPB1 (S78) alongside other signaling components and cellular structures.

    • This technique reveals spatial relationships between phosphorylated proteins and subcellular compartments, providing insights into the localization-dependent functions of HSPB1.

  • Integration with Functional Assays:

    • Couple phosphorylation measurements with downstream functional readouts (cytoskeletal reorganization, apoptosis resistance, protein aggregation) to establish cause-effect relationships.

    • This integrated approach connects signaling events to their biological consequences.

By implementing these multi-parameter approaches, researchers can contextualize HSPB1 S78 phosphorylation within the broader signaling network, generating more comprehensive and mechanistically informative datasets compared to single-parameter analyses.

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 2024 Thebiotek. All Rights Reserved.