HSPB1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

HSPB1 recombinant monoclonal antibodies are laboratory-engineered antibodies produced using DNA recombinant technology. Key steps include:

  • Immunization: Animals (e.g., rabbits or mice) are immunized with synthetic peptides or full-length human HSPB1 protein .

  • B Cell Isolation: Positive B cells are screened for specificity to HSPB1 epitopes .

  • Gene Cloning: Light and heavy chain genes are amplified via PCR and cloned into plasmid vectors .

  • Expression: Vectors are transfected into host cells (e.g., CHO or HEK293) for antibody production .

  • Purification: Antibodies are purified via affinity chromatography .

These antibodies exhibit high specificity for human HSPB1, with cross-reactivity reported in mouse and rat models .

Applications and Performance

HSPB1 recombinant monoclonal antibodies are validated for diverse applications:

ApplicationRecommended DilutionKey Findings
Western Blot (WB)1:500–1:5,000Detects endogenous HSPB1 at ~22–28 kDa; identifies phosphorylation at S78/S82 .
Immunohistochemistry (IHC)1:20–1:200Localizes HSPB1 in tumor tissues (e.g., prostate, breast) and stress-induced cells .
Flow Cytometry (FC)1:20–1:200Quantifies HSPB1 expression in HeLa cells under UV/TPA treatment .
Immunofluorescence (IF)1:20–1:200Visualizes HSPB1 in cytoskeletal structures and stress granules .
ELISAMeasures HSPB1 levels in serum, plasma, or cell supernatants .

3.1. Neurodegenerative Diseases

  • Charcot-Marie-Tooth Disease (CMT2F): HSPB1 mutations (e.g., R127W, S135F, R136W) disrupt oligomerization and chaperone activity, leading to axonal degeneration . Recombinant antibodies enable detection of hyperactive mutants with altered microtubule interactions .

  • Alzheimer’s Disease: HSPB1 sequesters Aβ oligomers into non-toxic aggregates, a mechanism validated in transgenic mouse models .

3.2. Cancer Biomarker

  • HSPB1 overexpression correlates with poor prognosis in melanoma, prostate, and breast cancers . Antibodies like clone CPTC-HSPB1-2 (ab268073) are used for tumor stratification .

3.3. Stress Response

  • HSPB1 translocates to the nucleus under stress, stabilizing actin and inhibiting caspase-mediated apoptosis . Phospho-specific antibodies (e.g., anti-S82) track activation dynamics .

Comparative Analysis of Select Clones

CloneVendorApplicationsPhospho-Specificity
CB2 (MAB21646)AbnovaWB, FCS82 phosphorylation
CPTC-HSPB1-2AbcamIF, IHC, FCPan-HSPB1
10C11EpigenTekWB, IHCN/A
66767-1-IgProteintechWB, IF, IHCDetects dimeric HSPB1

Challenges and Future Directions

  • Specificity: Some clones cross-react with HSPB6 (HSP20) in hetero-oligomer assays .

  • Therapeutic Potential: Antibodies targeting HSPB1’s ACD domain may modulate its chaperone activity in CMT2F .

References

  1. HSPB1 (phospho S82) monoclonal antibody, clone CB2 - Abnova .

  2. Production protocol for HSPB1 recombinant antibodies - Cusabio .

  3. Anti-HSPB1 antibody performance in IHC and WB - Biocompare .

  4. HSPB1 mutations and chaperone activity - Nature .

  5. Commercial HSPB1 ELISA kits - Sigma-Aldrich .

  6. HSPB1 gene and protein features - Thermo Fisher .

  7. HSPB1 in Alzheimer’s models - PubMed .

  8. Anti-Hsp27 antibody validation - Abcam .

  9. HSPB1 as a cancer marker - Proteintech .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch products within 1-3 business days after receiving your order. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery timeframes.
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, a small heat shock protein, acts as a molecular chaperone. Its primary function is to stabilize denatured proteins, keeping them in a state conducive to proper folding. HSPB1 plays a crucial role in stress resistance and actin organization. Through its molecular chaperone activity, HSPB1 likely regulates various biological processes, including the phosphorylation and axonal transport of neurofilament proteins.
Gene References Into Functions
  1. Research indicates that both the small heat shock protein HspB1/Hsp27 and the constitutive chaperone Hsc70/HspA8 interact with tau to prevent the formation of tau-fibrils and amyloid. HspB1 delays tau-fibril formation by weakly interacting with early aggregation intermediates. (HspB1 = heat shock protein family B small member 1; Hsc70 = heat shock protein family A Hsp70) PMID: 29298892
  2. HSP27 interacts with JAK2-STAT5 and is a potential therapeutic target in myelofibrosis. PMID: 29650953
  3. Studies have shown that HSP27 expression is significantly elevated in metastatic prostate cancer and serves as a strong prognostic marker for survival, alongside ALDH6A1 and prohibitin. PMID: 30396985
  4. High HSP27 expression is associated with breast cancer. PMID: 30106436
  5. Research suggests that overexpression of peroxiredoxin-2, annexin A2, and heat shock protein beta-1 correlates with tumor invasion, metastasis, and poor prognosis, suggesting their potential as diagnostic and therapeutic biomarkers. PMID: 29332450
  6. ETAS down-regulates HSP27 and pHSP27 (serine 78) in KLM1-R cells, while HSP70 and GRP78 levels remain unaffected. This research suggests that ETAS could enhance anticancer effects when combined with gemcitabine for patients with pancreatic cancer. PMID: 29936456
  7. Silencing 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 optimal ratio of Lipofectamine(R) 2000 and HSP27 plasmid. PMID: 29568951
  9. HSP27 expression in EUS-FNA samples appears to be useful for predicting sensitivity to GEM. PMID: 29695571
  10. High hsp27 expression is associated with leukemia. PMID: 29328466
  11. p-Hsp27 serves as a novel biomarker for glioma and may hold significant clinical value for further classifying patients with wild-type IDH1 and normal ATRX expression, as well as for evaluating prognosis. PMID: 29550762
  12. The dimer-monomer equilibrium of human HSP27 is influenced by the in-cell macromolecular crowding environment and is regulated 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. While the addition of apatorsen to chemotherapy did not improve outcomes in unselected patients with metastatic pancreatic cancer in the first-line setting, a trend towards prolonged PFS and OS in patients with high baseline serum Hsp27 suggests that this therapy may warrant further evaluation in this specific subgroup. PMID: 28935773
  16. High HSP27 expression is associated with metastasis in non-small cell lung cancer. PMID: 28656293
  17. Knockdown of HSP27 blocked TGF-beta-induced cisplatin resistance. PMID: 28848138
  18. HSP27 expression was approximately 2-fold higher in apical periodontitis. Additionally, increased HSP27 expression was detected in epithelial cells. PMID: 28673495
  19. Findings indicate the heat-shock protein 27 (Hsp27) pathway as a potential therapeutic target for managing conditions characterized by dysregulated interleukin 1 beta (IL-1beta) production. PMID: 27976724
  20. Increased protein levels of HSP27 through SUMO2/3-mediated SUMOylation play crucial roles in the progression of primary hepatocellular carcinoma. PMID: 28665748
  21. When examining the correlations between markers and the response to neoadjuvant chemotherapy, 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 valuable for further study in a larger patient population. PMID: 28608263
  22. Cetuximab and various concentrations of AG490, an inhibitor of JAK2, STAT3, and HSP27 protein levels, except in the KRAS G12V mutant line, SW620...cetuximab may enhance SN38 sensitivity by suppressing HSP27, through blocking the JAK/STAT signaling pathway, and exhibits synergistic effects when combined with SN38 in wild-type RAS CRC cells. PMID: 28656305
  23. Research concluded that HSP27-silenced placenta-derived multipotent cells differentiated into neurons exhibiting the characteristics of functional glutamatergic neurons. PMID: 27444754
  24. Data show that Hsp27 increases the degradation rate of ubiquitinated MST1, thus interrupting the Hippo pathway kinase cascade. Consequently, YAP and TAZ become less phosphorylated and free to translocate into the nucleus, promoting a malignant phenotype. These findings underscore the central importance 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 promote cholesterol efflux through activation of the PI3K/PKCzeta/Sp1 signal pathway in THP-1 macrophage-derived foam cells. PMID: 28610841
  28. This study presents 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 exhibits dynamic and complex rearrangements in response to changes in the cellular environment or when cell physiology is altered. [review] PMID: 28144778
  31. Glutathione-S-transferase - HspB1 fusion protein prevents a greater degree of malate dehydrogenase aggregation compared to glutathione-S-transferase -HspB5 fusion protein and wild type HspB1. PMID: 28130664
  32. Data suggest that oncogene-addicted cells rely on the small heat-shock protein of 27 kDa (HSP27) for survival and that HSP27 may interfere with the effectiveness of targeted agents. PMID: 28182330
  33. Computational drug repositioning has identified six cytostatic drugs that inhibit heat shock 27 kDa protein (HSP27) and address drug resistance. PMID: 27626687
  34. Distinct from the C-M-T phenotype observed in hereditary neuropathies caused by mutations in the HSPB1 gene. PMID: 27816334
  35. Knockdown of HSPB1 further increased the proportion of apoptotic cells in hyperthermic treated melanoma cells when 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 altering the dimerization of HSP27 presents a promising strategy for anticancer therapy in HSP27-overexpressing cancer cells. PMID: 27449291
  37. Apatorsen (a 2'-methoxyethyl-modified antisense oligonucleotide that inhibits Hsp27 expression) was well-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. These results highlight the central role of Hsp27 in RACK1 pseudosubstrate or LPS-induced cell activation of primary leukocytes. PMID: 27178349
  39. High p-Hsp27 expression is linked to 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. This 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, particularly in their 5' and 3'UTRs. These 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. Research suggests that HSPB6 forms hetero-oligomers with HSPB1 following these rules: (1) the highly conserved motif RLFDQXFG is essential for subunit exchange among oligomers, (2) a site approximately 20 residues downstream of this motif determines the size of the resulting hetero-oligomers, and (3) a region in the N-terminal domain 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 ability of sHsps to suppress the aggregation of model substrates is correlated with their capacity to form stable substrate complexes; notably, 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 highly effective inhibitors of alpha-syn aggregation. PMID: 27587396
  50. This study highlights a novel relationship between serum pHSP27 and BMD in Indian women; however, these findings require confirmation 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

What is HSPB1 and why is it significant in research?

HSPB1, also known as Heat Shock Protein 27 (HSP27), belongs to the small heat shock protein family (HSP20). In humans, the canonical protein consists of 205 amino acid residues with a molecular mass of 22.8 kDa and localizes to both the nucleus and cytoplasm . This protein is widely expressed across multiple tissue types and plays crucial roles in intracellular signal transduction and regulation of apoptosis .

HSPB1 is particularly significant in research due to its involvement in cellular stress responses, protein folding, and cytoprotection. Additionally, its altered expression has been implicated in various pathological conditions including neurodegenerative disorders, cardiovascular diseases, and cancer progression. The protein undergoes important post-translational modifications, notably phosphorylation at several serine residues (including S15, S78, S82, and S83), which regulate its functional properties .

Orthologs of HSPB1 have been identified across multiple species including mouse, rat, bovine, frog, zebrafish, chimpanzee, and chicken, making it a valuable target for comparative studies of cellular stress responses across evolutionary lineages .

What detection methods work best with HSPB1 recombinant monoclonal antibodies?

HSPB1 recombinant monoclonal antibodies are versatile tools compatible with multiple detection methods. Western Blot (WB) represents the most widely used application, allowing precise quantification of total and phosphorylated HSPB1 levels in various sample types .

For cellular localization studies, Immunocytochemistry (ICC), Immunofluorescence (IF), and Immunohistochemistry (IHC) techniques offer excellent visualization of HSPB1 distribution patterns . Many commercially available antibodies are validated for these applications, with some specifically optimized for paraffin-embedded tissue sections (IHC-p) .

Additional compatible methodologies include:

  • Flow Cytometry (FCM) for single-cell analyses

  • Enzyme-Linked Immunosorbent Assay (ELISA) for quantitative measurement

  • Immunoprecipitation (IP) for protein-protein interaction studies

When selecting an antibody, researchers should prioritize products with validation data specifically for their intended application and target species. Cross-reactivity profiles vary significantly between antibodies, with many validated for human HSPB1 detection, while others demonstrate broader reactivity across multiple species (human, mouse, rat) .

How should HSPB1 recombinant monoclonal antibodies be stored and handled?

Proper storage and handling of HSPB1 recombinant monoclonal antibodies are essential for maintaining their performance and specificity. Most commercial antibodies should be stored at -20°C for long-term preservation, though specific recommendations may vary by manufacturer .

For working solutions, aliquoting the stock antibody into smaller volumes is strongly recommended to minimize freeze-thaw cycles, which can degrade antibody quality. These aliquots should be prepared using sterile techniques and stored in appropriate buffer conditions (typically PBS with a stabilizing protein such as BSA and preservatives like sodium azide).

During experimental procedures:

  • Allow antibodies to equilibrate to room temperature before opening to prevent condensation

  • Maintain sterile handling procedures to prevent contamination

  • Use appropriate dilution buffers as recommended by the manufacturer

  • Prepare working dilutions immediately before use for optimal performance

For phospho-specific HSPB1 antibodies (targeting sites like S15, S28, S78, S82), additional precautions may be necessary, including the use of phosphatase inhibitors in sample preparation to preserve the phosphorylation status of the target epitope .

What dilution ranges are typically effective for HSPB1 antibodies in different applications?

Optimal dilution ranges for HSPB1 antibodies vary significantly depending on the specific application, antibody affinity, and detection system employed. Based on available product information, the following ranges represent starting points for optimization:

ApplicationTypical Dilution RangeNotes
Western Blot1:500 - 1:5000Higher dilutions (1:2000+) often work with high-affinity monoclonal antibodies
Immunohistochemistry1:50 - 1:500Paraffin sections may require additional optimization
Immunofluorescence1:100 - 1:1000Signal intensity varies with fixation method
ELISA1:1000 - 1:10000Sandwich ELISA typically requires matched antibody pairs
Flow Cytometry1:50 - 1:200Cell permeabilization required for intracellular staining

Each new lot of antibody should undergo titration to determine the optimal working concentration for specific experimental conditions . When switching between applications, re-optimization is strongly recommended rather than simply converting dilution factors.

How do I design experiments to distinguish between different phosphorylated forms of HSPB1?

HSPB1 undergoes complex regulation through phosphorylation at multiple serine residues, primarily S15, S78, S82, and S83, each associated with distinct functional outcomes. Designing experiments to differentiate between these phosphorylation states requires careful planning and specialized reagents.

Phospho-specific antibodies targeting individual phosphorylation sites (such as S78 or S82) provide the most direct approach . Several commercial sources offer such antibodies with high specificity. To ensure robust results:

  • Include appropriate controls:

    • Unphosphorylated recombinant HSPB1 protein

    • Samples treated with lambda phosphatase to remove phosphorylation

    • Positive controls where phosphorylation is induced (e.g., heat shock, oxidative stress)

  • Consider multidimensional approaches:

    • Combine Western blotting with phospho-specific antibodies

    • Verify with mass spectrometry to identify and quantify specific phosphorylation sites

    • Use 2D gel electrophoresis to separate phospho-isoforms based on charge differences

  • For temporal dynamics studies:

    • Design time-course experiments following stimulation

    • Use phospho-specific antibodies in parallel with total HSPB1 detection

    • Calculate phospho/total HSPB1 ratios to normalize for expression level variations

When interpreting results, remember that different phosphorylation sites may have synergistic or antagonistic effects, and their biological significance can be cell-type and context-dependent.

What are the critical considerations for quantitative analysis of HSPB1 in different subcellular compartments?

HSPB1 exhibits dynamic subcellular localization patterns that change in response to various cellular stresses. Accurate quantitative analysis of HSPB1 distribution across subcellular compartments presents several methodological challenges that require careful consideration.

For immunofluorescence-based approaches:

  • Optimization of fixation methods is critical - paraformaldehyde typically preserves HSPB1 epitopes while maintaining cellular architecture

  • Include co-staining with established subcellular markers (nucleolin for nucleoli, DAPI for nucleus, phalloidin for actin cytoskeleton)

  • Implement rigorous image acquisition parameters:

    • Fixed exposure settings across samples

    • Z-stack acquisition to capture the full cellular volume

    • Multichannel acquisition with minimal bleed-through

For biochemical fractionation approaches:

  • Validate the purity of subcellular fractions using compartment-specific markers

  • Account for potential cross-contamination between fractions

  • Normalize HSPB1 levels to total protein content in each fraction

Advanced quantification strategies include:

  • Digital image analysis with automated compartment segmentation

  • Calculation of nuclear/cytoplasmic ratios through pixel intensity measurements

  • Correlation coefficient analysis for co-localization with other proteins of interest

The translocation of HSPB1 between compartments often correlates with its phosphorylation status, with certain phospho-forms showing preferential localization patterns. Therefore, combining phospho-specific antibodies with subcellular localization studies can provide mechanistic insights into HSPB1 regulation .

How can researchers troubleshoot non-specific binding or weak signals when using HSPB1 antibodies?

Non-specific binding and weak signal issues represent common challenges when working with HSPB1 antibodies. Systematic troubleshooting approaches can help identify and resolve these technical problems.

For non-specific binding issues:

  • Optimize blocking conditions:

    • Test different blocking agents (BSA, non-fat milk, normal serum)

    • Increase blocking time or concentration

    • Consider specialized blocking reagents for problematic samples

  • Adjust antibody conditions:

    • Titrate antibody concentration to find optimal signal-to-noise ratio

    • Increase washing stringency (longer washes, higher detergent concentration)

    • Perform overnight incubations at 4°C rather than shorter incubations at room temperature

  • Validate antibody specificity:

    • Include HSPB1 knockout/knockdown controls

    • Pre-absorb antibody with recombinant antigen

    • Compare results from multiple antibodies targeting different epitopes

For weak signal issues:

  • Sample preparation adjustments:

    • Optimize protein extraction methods to maximize HSPB1 recovery

    • Add protease and phosphatase inhibitors during sample preparation

    • Avoid freeze-thaw cycles of protein samples

  • Detection system enhancements:

    • Switch to more sensitive detection systems (e.g., chemiluminescence to ECL-Plus)

    • Consider signal amplification methods (biotin-streptavidin systems)

    • For immunofluorescence, try tyramide signal amplification

  • Antibody selection considerations:

    • Monoclonal antibodies typically offer higher specificity but may have lower sensitivity

    • Polyclonal antibodies generally provide stronger signals but potentially more background

    • Recombinant antibodies offer advantages in batch-to-batch consistency

A systematic approach to troubleshooting, changing one variable at a time, facilitates identification of the specific factors affecting antibody performance in your experimental system.

What methodological approaches enable accurate comparison of HSPB1 levels across different experimental conditions?

Accurate comparison of HSPB1 levels across experimental conditions requires rigorous methodological standardization and appropriate normalization strategies to minimize technical variability and ensure biological significance.

For Western blot quantification:

  • Implementation of standardized protocols:

    • Load equal protein amounts confirmed by BCA/Bradford assay

    • Include gradient standards of recombinant HSPB1 for calibration curves

    • Process all samples simultaneously when possible

  • Normalization strategies:

    • Use multiple housekeeping proteins (β-actin, GAPDH, tubulin) as loading controls

    • Calculate relative expression ratios (HSPB1/loading control)

    • Consider total protein normalization via stain-free technology or Ponceau staining

  • Image acquisition considerations:

    • Ensure signal is within linear detection range of imaging system

    • Avoid saturated pixels which invalidate quantitative analysis

    • Use consistent exposure settings across comparable blots

For ELISA-based quantification:

  • Standard curve preparation:

    • Use recombinant HSPB1 calibrators covering the expected concentration range

    • Process standards and samples in the same plate

    • Implement appropriate curve-fitting models (4PL or 5PL)

  • Quality control measures:

    • Include internal control samples across multiple plates

    • Calculate intra- and inter-assay coefficients of variation

    • Validate dilution linearity to confirm assay reliability

Advanced considerations include:

  • For phosphorylated HSPB1 analysis, calculate phospho/total ratios to distinguish regulation by phosphorylation from changes in expression

  • When comparing across cell types or tissues with different baseline expression, consider fold-change relative to appropriate controls

  • For time-course experiments, analyze area under the curve rather than individual time points

Statistical analysis should incorporate appropriate tests for multiple comparisons when analyzing HSPB1 levels across diverse experimental conditions.

What are the critical cellular pathways and functions mediated by HSPB1?

HSPB1 functions as a multifaceted molecular chaperone with diverse roles in cellular homeostasis and stress response pathways. Its involvement spans numerous critical biological processes:

  • Stress response and cytoprotection:

    • Prevents protein aggregation during cellular stress

    • Stabilizes cytoskeletal elements (particularly actin filaments)

    • Inhibits apoptosis through multiple mechanisms including interaction with cytochrome c and procaspase-3

    • Enhances cellular antioxidant capacity by maintaining glutathione in reduced form

  • Signal transduction pathways:

    • Modulates p38 MAPK signaling through direct interactions

    • Influences NF-κB activation with context-dependent effects

    • Regulates TGF-β/Smad signaling in fibrotic responses

    • Participates in IGF-1 pathway regulation in some cell types

  • Protein quality control:

    • Facilitates proper protein folding as a molecular chaperone

    • Contributes to targeting misfolded proteins for degradation

    • Interacts with the ubiquitin-proteasome system components

    • Stabilizes nascent proteins during translation

The functional versatility of HSPB1 is regulated primarily through its phosphorylation status, which influences its oligomerization state and subsequent molecular interactions. Under basal conditions, HSPB1 typically exists as large oligomers (up to 800 kDa), while stress-induced phosphorylation promotes dissociation into smaller units with distinct functional properties .

Understanding these pathways provides critical context for interpreting experimental results involving HSPB1 detection and manipulation.

How can researchers effectively study the relationship between HSPB1 phosphorylation and its cellular functions?

Investigating the complex relationship between HSPB1 phosphorylation and its diverse cellular functions requires multifaceted experimental approaches that combine detection methods with functional assays.

Experimental strategies to establish phosphorylation-function relationships include:

  • Phosphomimetic and phospho-null mutants:

    • Generate HSPB1 constructs with serine-to-aspartate (phosphomimetic) or serine-to-alanine (phospho-null) mutations at specific sites (S15, S78, S82, S83)

    • Express these constructs in HSPB1-depleted backgrounds to assess functional rescue

    • Compare cellular outcomes including stress resistance, apoptosis susceptibility, and protein aggregation

  • Pharmacological manipulation:

    • Employ selective inhibitors of kinases responsible for HSPB1 phosphorylation (p38 MAPK, MAPKAPK-2, PKC, PKD)

    • Use phosphatase inhibitors to maintain phosphorylation status

    • Monitor functional consequences while confirming phosphorylation status using phospho-specific antibodies

  • Time-resolved analyses:

    • Track HSPB1 phosphorylation kinetics following stress induction

    • Correlate phosphorylation patterns with functional outcomes at each time point

    • Implement mathematical modeling to infer causal relationships

Phosphorylation SitePrimary KinaseFunctional ImpactDetection Method
Ser15MAPKAPK-2, MAPKAPK-3Regulates oligomerization, cytoskeletal stabilizationPhospho-specific antibodies, phosphoproteomics
Ser78MAPKAPK-2, MAPKAPK-3Modulates chaperone activity, anti-apoptotic functionPhospho-specific antibodies, 2D gel electrophoresis
Ser82MAPKAPK-2, MAPKAPK-3, PKC, PKDCritical for actin regulation, stress granule associationPhospho-specific antibodies, in vitro kinase assays
Ser83PKA, PKC, PKGFine-tunes protein-protein interactionsPhospho-specific antibodies, mass spectrometry

When interpreting results, researchers should consider that different phosphorylation sites may have synergistic or antagonistic effects, and their biological significance often depends on cellular context, stress type, and temporal dynamics .

What considerations are important when studying HSPB1 in disease models and potential therapeutic applications?

HSPB1 has been implicated in numerous pathological conditions, making it a valuable target for disease model development and therapeutic intervention studies. Researchers investigating HSPB1 in disease contexts should consider several critical factors:

  • Disease-specific expression patterns:

    • HSPB1 shows altered expression and phosphorylation in many diseases

    • Cancer: typically upregulated, associated with treatment resistance

    • Neurodegenerative disorders: found in protein aggregates in ALS, Alzheimer's

    • Cardiovascular diseases: cardioprotective in ischemia models

    • Inflammatory conditions: context-dependent pro- or anti-inflammatory effects

  • Model system selection considerations:

    • Cell culture models: consider endogenous HSPB1 expression levels

    • Animal models: note species-specific differences in HSPB1 regulation

    • Patient-derived samples: standardize collection and processing protocols

    • Disease-specific stimuli: employ relevant stressors (oxidative stress, hypoxia, etc.)

  • Therapeutic targeting approaches:

    • Direct HSPB1 modulation: overexpression, siRNA/shRNA knockdown

    • Indirect regulation: kinase inhibitors affecting phosphorylation status

    • Peptide-based approaches: targeting specific HSPB1 interaction domains

    • Delivery challenges: tissue-specific targeting strategies

When designing therapeutic intervention studies, researchers should implement appropriate controls, including:

  • Vehicle-only treatments

  • Non-targeting sequences for RNA interference

  • Scrambled peptides for protein-protein interaction inhibitors

  • Wild-type HSPB1 expression for comparison with mutant forms

For translational research, it's essential to validate findings across multiple model systems and correlate results with clinical data when available. The phosphorylation status of HSPB1 should be monitored alongside total protein levels, as therapeutic approaches may differentially affect specific phospho-forms with distinct functional properties .

How does HSPB1 interact with other heat shock proteins and molecular chaperones?

HSPB1 functions within a complex network of heat shock proteins and molecular chaperones, engaging in both physical interactions and functional cooperation to maintain cellular proteostasis. Understanding these interactions provides critical context for experimental design and data interpretation.

Key interaction partners and networks include:

  • Interactions with other small heat shock proteins:

    • Forms heterooligomeric complexes with other sHSPs including HSPB5 (αB-crystallin) and HSPB6

    • Heterooligomerization modifies chaperone activity and substrate specificity

    • Phosphorylation status influences these interaction dynamics

    • Co-expression patterns vary by tissue type and stress conditions

  • Functional cooperation with ATP-dependent chaperones:

    • Acts as a "holdase" to prevent irreversible protein aggregation

    • Transfers partially folded clients to HSP70/HSP40 system for ATP-dependent refolding

    • Collaborates with HSP90 in stabilization of specific client proteins

    • Participates in triage decisions between protein refolding and degradation pathways

  • Interactions with cytoskeletal components:

    • Directly binds to actin filaments, modulating polymerization dynamics

    • Stabilizes intermediate filaments through phosphorylation-dependent interactions

    • Protects cytoskeletal integrity during cellular stress

    • Influences cell migration through cytoskeletal regulation

Experimental approaches to study these interactions include:

  • Co-immunoprecipitation with HSPB1-specific antibodies followed by mass spectrometry

  • Proximity ligation assays for in situ detection of protein-protein interactions

  • FRET-based approaches to monitor interaction dynamics in living cells

  • Split-protein complementation assays for validation of direct interactions

When designing experiments to investigate HSPB1 interactions, researchers should consider that:

  • Interaction patterns change dramatically under stress conditions

  • Phosphorylation significantly alters HSPB1's interactome

  • Cell type-specific factors influence chaperone network composition

  • Temporal dynamics are critical, as interactions may be transient

Understanding these complex interaction networks provides essential context for interpreting experimental results and developing targeted intervention strategies in various disease states .

What are the key differences between polyclonal, monoclonal, and recombinant antibodies for HSPB1 detection?

Researchers have multiple antibody types available for HSPB1 detection, each with distinct characteristics that influence their performance in specific applications. Understanding these differences is essential for selecting the optimal reagent for particular experimental needs.

Antibody TypeProduction MethodAdvantagesLimitationsBest Applications
PolyclonalImmunization of animals (typically rabbits) with HSPB1 antigen- Recognizes multiple epitopes
- Higher sensitivity
- More tolerant of minor protein modifications
- Batch-to-batch variation
- Higher background potential
- Limited supply
- Western blot for maximum sensitivity
- IHC of fixed tissues
- Applications where sensitivity outweighs specificity
MonoclonalSingle B-cell clone producing identical antibodies- Consistent reproducibility
- High specificity for single epitope
- Lower background
- Unlimited supply
- May be sensitive to epitope modifications
- Generally lower affinity
- May be sensitive to denaturing conditions
- Flow cytometry
- Quantitative applications requiring consistency
- Detection of specific isoforms
Recombinant MonoclonalCloned antibody genes expressed in defined expression systems- Exceptional batch-to-batch consistency
- Defined sequence
- No animal use
- Potential for engineering
- Higher cost
- More limited selection
- Newer technology with less validation history
- Critical quantitative applications
- Phospho-specific detection
- Long-term studies requiring consistent reagents

For HSPB1 applications specifically:

  • Polyclonal antibodies often provide stronger signals in Western blot and IHC applications, making them valuable for detecting low expression levels

  • Monoclonal antibodies are preferred for distinguishing between HSPB1 and closely related heat shock protein family members due to their specificity

  • Recombinant monoclonal antibodies offer the highest consistency for quantitative applications and longitudinal studies

When selecting an antibody, researchers should also consider the specific epitope targeted, especially for phospho-specific detection or when studying particular domains of HSPB1 with distinct functions.

How should researchers validate HSPB1 antibodies for their specific experimental systems?

Thorough validation of HSPB1 antibodies in the specific experimental system is essential for generating reliable and reproducible results. A comprehensive validation strategy should include multiple approaches:

  • Positive and negative controls:

    • Positive: Samples with known HSPB1 expression (heat-shocked cells, tissues with high endogenous expression)

    • Negative: HSPB1 knockout/knockdown samples, tissues known not to express HSPB1

    • Recombinant HSPB1 protein as standard for size verification

  • Specificity confirmation:

    • Western blot showing single band at expected molecular weight (approximately 22.8 kDa)

    • Pre-absorption with recombinant antigen to confirm specific binding

    • Comparison of results from multiple antibodies targeting different epitopes

    • Peptide competition assays to verify epitope specificity

  • Application-specific validation:

    • For Western blot: Confirm linear dynamic range using dilution series

    • For ICC/IF: Verify subcellular localization patterns match known distribution

    • For IHC: Compare staining patterns with published literature and validate preservation of epitopes in fixed tissues

    • For phospho-specific antibodies: Confirm detection is eliminated by phosphatase treatment

  • Cross-reactivity assessment:

    • Test against closely related family members (other small HSPs)

    • Verify species cross-reactivity for studies involving multiple organisms

    • Check for non-specific binding in complex samples

Researchers should document validation results thoroughly and consider publishing validation data as supplementary material in publications to enhance reproducibility. For phospho-specific HSPB1 antibodies, additional validation should confirm specificity for the particular phosphorylation site rather than just recognition of phosphorylated proteins in general .

What are the most effective protocols for detecting phosphorylated HSPB1 in various sample types?

Detecting phosphorylated forms of HSPB1 presents unique challenges that require specialized protocols optimized for preserving and accurately measuring these transient modifications. The effectiveness of phospho-HSPB1 detection depends significantly on sample preparation and detection methodology.

For cell and tissue lysate preparation:

  • Rapid sample collection and processing to prevent dephosphorylation

  • Inclusion of robust phosphatase inhibitor cocktails in all buffers

  • Maintenance of cold temperatures throughout processing

  • Consideration of specialized lysis buffers optimized for phosphoprotein preservation

Western blot optimization for phospho-HSPB1:

  • Preferential use of PVDF membranes which typically provide better retention of phosphoproteins

  • Implementation of phospho-specific blocking buffers (containing phosphatase inhibitors)

  • Extended primary antibody incubation (overnight at 4°C) for optimal binding

  • Selection of sensitive detection systems (enhanced chemiluminescence or fluorescent secondaries)

For immunohistochemical detection:

  • Optimization of fixation protocols that preserve phospho-epitopes

  • Consideration of antigen retrieval methods specifically validated for phosphoproteins

  • Inclusion of phosphatase inhibitors in staining buffers

  • Use of amplification systems for detecting low-abundance phospho-forms

Verification approaches include:

  • Parallel detection of total HSPB1 to calculate phosphorylation ratios

  • Treatment of control samples with lambda phosphatase to confirm specificity

  • Inclusion of positive controls with enhanced phosphorylation (heat shock, oxidative stress)

  • Correlation with known physiological/pathological states affecting HSPB1 phosphorylation

For phospho-specific sites (S15, S78, S82, S83), researchers should be aware that different sites may require specific optimization, as antibody accessibility and phosphorylation dynamics vary between sites .

What are the critical considerations for multiplex detection of HSPB1 with other proteins of interest?

For fluorescence-based multiplex immunodetection:

  • Antibody selection considerations:

    • Choose primary antibodies from different host species to avoid cross-reactivity

    • For same-species antibodies, use directly conjugated primaries or sequential detection protocols

    • Verify spectral separation of fluorophores to minimize bleed-through

    • Consider antibody formats (Fab fragments, nanobodies) for dense epitope regions

  • Protocol optimization:

    • Titrate each antibody individually before combining in multiplex

    • Implement appropriate blocking strategies to minimize background

    • Include single-stained controls for accurate compensation

    • Consider tyramide signal amplification for low-abundance targets

  • Imaging and quantification:

    • Acquire appropriate single-stained controls for spectral unmixing

    • Use consistent exposure settings across experimental groups

    • Implement automated analysis workflows for unbiased quantification

    • Consider colocalization analyses when appropriate

For multiplex Western blotting:

  • Sample preparation considerations:

    • Ensure extraction conditions are compatible with all target proteins

    • Consider molecular weight differences for multiplexing strategy (stripping vs. same blot)

    • Optimize loading to accommodate different abundance levels

  • Technical approaches:

    • Traditional sequential stripping and reprobing (risk of protein loss)

    • Spectrally distinct fluorescent secondaries for simultaneous detection

    • Different visualization methods (chemiluminescence vs. fluorescence)

When multiplexing HSPB1 with phosphorylated forms:

  • Be aware of potential epitope masking issues

  • Consider the use of phospho-specific and total protein antibodies from different species

  • Validate that detection of one form doesn't interfere with the other

Spatial considerations for tissue analysis include:

  • Optimization of antigen retrieval conditions compatible with all targets

  • Sequential detection protocols when necessary

  • Digital slide scanning with multispectral capabilities for comprehensive analysis

What emerging technologies are advancing HSPB1 research?

HSPB1 research continues to evolve with the integration of cutting-edge technologies that enhance detection sensitivity, spatial resolution, and functional analysis capabilities. These emerging approaches are expanding our understanding of HSPB1 biology in normal and pathological states.

Single-cell technologies have revolutionized the field by revealing heterogeneity in HSPB1 expression and phosphorylation patterns within seemingly homogeneous populations. Single-cell RNA sequencing and mass cytometry provide unprecedented resolution of HSPB1 regulation across diverse cell types and states, while newer techniques like spatial transcriptomics maintain tissue context.

Advanced imaging approaches are transforming our ability to visualize HSPB1 dynamics:

  • Super-resolution microscopy (STORM, PALM, STED) reveals HSPB1 distribution at nanometer scale

  • Live-cell imaging with genetically encoded fluorescent tags captures real-time HSPB1 translocation

  • Fluorescence correlation spectroscopy analyzes HSPB1 oligomerization states in living cells

  • Lattice light-sheet microscopy provides extended imaging of HSPB1 dynamics with minimal phototoxicity

Proteomics advances have enhanced our understanding of HSPB1 interactions and modifications:

  • Proximity labeling methods (BioID, APEX) identify transient HSPB1 interaction partners

  • Crosslinking mass spectrometry reveals structural details of HSPB1 complexes

  • Targeted proteomics approaches enable precise quantification of specific HSPB1 phospho-forms

  • Thermal proteome profiling detects HSPB1 stabilization during stress responses

CRISPR-based technologies offer new approaches for HSPB1 functional studies:

  • Base editing for introduction of specific phospho-null or phosphomimetic mutations

  • CRISPRi/CRISPRa for modulation of HSPB1 expression levels

  • CRISPR screens to identify genetic interactions with HSPB1

These technological advances are generating more comprehensive datasets that, when integrated through computational approaches, provide systems-level insights into HSPB1 function across diverse biological contexts .

How can researchers contribute to improving reproducibility in HSPB1 antibody-based research?

Enhancing reproducibility in HSPB1 antibody-based research requires conscious implementation of best practices throughout the experimental workflow. Researchers can make significant contributions to field-wide reproducibility through several key strategies.

Comprehensive antibody validation and documentation:

  • Perform and report thorough validation using multiple methods (Western blot, immunoprecipitation, knockout controls)

  • Document full antibody information in publications (supplier, catalog number, lot number, RRID)

  • Include validation data as supplementary material in publications

  • Contribute validation data to community resources and antibody validation databases

Rigorous experimental design:

  • Include appropriate positive and negative controls in every experiment

  • Implement blinding procedures for analysis where feasible

  • Determine sample sizes through power analysis

  • Pre-register experimental protocols when possible

Detailed methodological reporting:

  • Provide complete protocol details including:

    • Sample preparation methods (lysis buffers, inhibitors used)

    • Antibody concentrations and incubation conditions

    • Detection systems and image acquisition parameters

    • Analysis methods including software and parameters

  • Consider protocol publication in dedicated journals or repositories

  • Make raw data available through appropriate repositories

  • Implement version control for analysis scripts and processing workflows

Standardization efforts:

  • Adopt community standards for antibody-based techniques when available

  • Participate in multi-laboratory validation studies

  • Utilize reference materials and standards when possible

  • Consider implementing automated protocols to reduce technical variation

Selection of appropriate antibody formats:

  • Consider the advantages of recombinant monoclonal antibodies for long-term reproducibility

  • For critical measurements, validate findings with multiple antibodies targeting different epitopes

  • Be aware of lot-to-lot variation, particularly with polyclonal antibodies

  • When feasible, validate commercial findings with independent development of research antibodies

These collective efforts will strengthen the reliability of HSPB1 research findings and accelerate progress in understanding this important protein's roles in health and disease.

What are the most promising directions for future HSPB1 functional studies?

Future HSPB1 research holds significant promise for advancing our understanding of stress response mechanisms and developing novel therapeutic approaches. Several directions appear particularly promising for functional studies:

Structural biology approaches:

  • Cryo-electron microscopy of HSPB1 oligomeric complexes to resolve dynamic structural states

  • Hydrogen-deuterium exchange mass spectrometry to map conformational changes during stress

  • NMR studies of phosphorylation-induced structural rearrangements

  • Computational modeling of HSPB1 interactions with client proteins

Cell-type specific functions:

  • Single-cell approaches to resolve HSPB1 roles across heterogeneous populations

  • Tissue-specific conditional knockout models to address developmental compensation

  • Cell type-specific phosphorylation patterns and their functional consequences

  • Specialized functions in stem cells versus differentiated populations

Mechanistic studies of HSPB1 in emerging contexts:

  • Roles in liquid-liquid phase separation and biomolecular condensate regulation

  • Functions in RNA metabolism and stress granule dynamics

  • Contributions to mitochondrial quality control and metabolic adaptation

  • Involvement in intercellular communication via extracellular vesicles

Translational research directions:

  • Development of small molecule modulators of HSPB1 oligomerization

  • Phosphorylation site-specific therapeutic targeting

  • HSPB1-based biomarkers for stress response activation in pathological states

  • Combinatorial approaches targeting multiple heat shock proteins simultaneously

Integration of HSPB1 within systems biology frameworks:

  • Network analysis of HSPB1 interactome across stress conditions

  • Mathematical modeling of HSPB1 phosphorylation dynamics

  • Multi-omics approaches to position HSPB1 within global stress responses

  • Evolutionary analysis of HSPB1 functions across species

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.