Phospho-CDC37 (S13) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism

CDC37 is a molecular chaperone that binds to Hsp90 and facilitates the maturation and stabilization of client kinases. Phosphorylation at S13 by casein kinase II (CK2) is essential for CDC37’s function, enhancing its interaction with Hsp90 and enabling nucleotide-driven conformational changes critical for kinase activation .

Key FeatureDescription
TargetPhosphorylated serine 13 (S13) of CDC37
FunctionDetects CDC37’s active state, enabling studies of kinase regulation and Hsp90 dynamics
HostRabbit (commonly recombinant)
ApplicationsWestern blot (WB), ELISA, immunohistochemistry (IHC), flow cytometry

Validation and Specificity

Antibodies are rigorously tested for specificity and cross-reactivity:

Western Blot Validation

  • Abcam’s EPR4879: Detects a 44 kDa band in HeLa lysates, absent when treated with phosphatase .

  • Cell Signaling’s D8P8F: Recognizes phosphorylated CDC37 in HeLa cells, with no signal in unphosphorylated controls .

  • Boster’s A02169S13-1: Blocks binding with non-phosphorylated peptides, confirming specificity .

Immunohistochemistry (IHC)

  • Boster’s A02169S13-1: Stains paraffin-embedded human lung cancer and placental tissues, with peptide blocking eliminating signal .

Functional Insights

  • Phosphorylation Independence in Viral Interactions: CDC37 stabilizes the rabies virus phosphoprotein (P) independently of S13 phosphorylation, suggesting novel chaperone mechanisms .

Cancer and Kinase Regulation

  • Kinase Stabilization: Phospho-CDC37 antibodies track Hsp90’s role in maintaining oncogenic kinases (e.g., CDK4, SRC) .

  • Therapeutic Targeting: Inhibiting CDC37-Hsp90 interactions disrupts kinase activity, offering potential cancer therapies .

Viral Pathogenesis

  • Rabies Virus: CDC37 binds the non-kinase P protein, stabilizing it during replication. Phosphorylation at S13 is not required for this interaction, highlighting divergent chaperone mechanisms .

Study FocusKey FindingsSource
Rabies Virus P ProteinCDC37 stabilizes P via Hsp90; S13 phosphorylation is dispensable for binding
CK2-Dependent PhosphorylationCK2 phosphorylates S13, enabling CDC37-Hsp90 interaction and kinase activation

Optimal Experimental Conditions

ParameterRecommendationSource
WB Dilution1:1,000–1:10,000
IHC Dilution1:100–1:300
Blocking Buffer5% NFDM/TBST or BSA
Storage-20°C (long-term), 4°C (short-term)

Limitations

  • Species Cross-Reactivity: Most antibodies target human, mouse, and rat CDC37, with limited data on other species .

  • Phosphatase Sensitivity: Signal loss post-phosphatase treatment confirms specificity but requires careful handling .

Product Specs

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

CUSABIO has engineered vector clones for the expression of a recombinant CDC37 antibody in mammalian cells. These vector clones were constructed by integrating the CDC37 antibody heavy and light chains into suitable expression vectors. The recombinant CDC37 antibody was subsequently purified from the culture medium using an affinity chromatography technique. This antibody can be utilized to detect CDC37 protein from human samples in ELISA and Western blotting applications.

The phospho-CDC37 (S13) antibody exhibits specificity towards phosphorylated CDC37. CDC37's functionality is regulated through phosphorylation at S13 by protein kinase CK2. This phosphorylation event at S13 is crucial for CDC37's kinase binding activity and its ability to facilitate the nucleotide-regulated conformational switching of Hsp90. CDC37 effectively suppresses the ATP hydrolysis step, enabling prolonged association of Hsp90 dimers with client proteins, leading to enhanced chaperone activity.

Form
Liquid
Lead Time
Generally, we can ship your orders within 1-3 business days of receipt. The delivery time may vary depending on the chosen shipping method and destination. For precise delivery timelines, please consult your local distributors.
Synonyms
CDC 37 antibody; Cdc37 antibody; CDC37 cell division cycle 37 homolog antibody; CDC37 cell division cycle 37 S cerevisiae homolog antibody; CDC37 cell division cycle 37; S cerevisiae; homolog of antibody; Cdc37 homolog antibody; CDC37 protein antibody; CDC37_HUMAN antibody; CDC37A antibody; cell division cycle 37 antibody; Cell division cycle 37 homolog antibody; Hsp90 chaperone protein kinase targeting subunit antibody; Hsp90 chaperone protein kinase targeting subunit p50Cdc37 antibody; Hsp90 chaperone protein kinase-targeting subunit antibody; Hsp90 co chaperone Cdc37 antibody; Hsp90 co-chaperone Cdc37 antibody; p50 antibody; p50Cdc37 antibody; S cerevisiae hypothetical protein CDC37 antibody
Target Names
Uniprot No.

Target Background

Function
CDC37 functions as a co-chaperone that binds to numerous kinases. This interaction facilitates the association of kinases with the Hsp90 complex, resulting in stabilization and enhanced activity of these kinases. Notably, CDC37 also inhibits the ATPase activity of HSP90AA1.
Gene References Into Functions
  1. Research has demonstrated that the Cdc37 gene is upregulated in human colorectal adenocarcinoma (CRC). Moreover, knockdown of Cdc37 effectively reduces cell proliferation, enhances apoptosis, and inhibits G1-S transition in CRC cells, whereas overexpression exhibits the opposite effects. Mechanistically, Cdc37 enhances CDK4 stability, promoting phosphorylation of RB1 and ultimately accelerating CRC progression. PMID: 29288563
  2. Within the kinase chaperone cycle, Cdc37, phosphorylated at Y298, acts as a docking site for non-receptor tyrosine kinases through their regulatory domains. This interaction drives the coupled phosphorylation of Hsp90 at Y197, specifically regulating kinase chaperoning. PMID: 29343704
  3. Findings suggest that this mechanism could be exploited by the Hsp90-Cdc37 chaperone system to recruit and protect inherently dynamic kinase clients from degradation. PMID: 29267381
  4. These results suggest a re-evaluation of Cdc37's role in the kinase lifecycle, indicating that such interactions potentially allow kinases to respond more swiftly to key signals while simultaneously protecting unstable kinases from degradation and suppressing unwanted basal activity. PMID: 28784328
  5. Niclosamide ethanolamine disrupts the interaction between cell division cycle 37 and heat shock protein 90 in hepatocellular carcinoma, leading to reduced tumor growth. PMID: 28284560
  6. Cdc37 performs a quality control function for protein kinases, including b-raf. In this context, induced conformational instability serves as a signal for Hsp90 dependence and stable cochaperone association. PMID: 27105117
  7. Ulk1 promotes the degradation of Hsp90-Cdc37 client kinases, resulting in increased cellular sensitivity to Hsp90 inhibitors. Consequently, this study provides evidence for an anti-proliferative role of Ulk1 in response to Hsp90 inhibition in cancer cells. PMID: 28073914
  8. The study indicates that the interaction between sB-Raf and the Hsp90 chaperone system is mediated by contacts with the M domain of Hsp90. This interaction contributes to the formation of the ternary complex with Cdc37 as long as the kinase is not stabilized by nucleotide. PMID: 27620500
  9. Beyond these distinct Cdc37/Hsp90 interfaces, the binding of the B-Raf protein kinase to the cochaperone is conserved across mammals and nematodes. PMID: 26511315
  10. Suppression of the cochaperone CDC37 expression in hepatocellular carcinoma cells inhibits cell cycle progression and cell growth. PMID: 25098386
  11. Correlation between PDZK1, Cdc37, Akt, and breast cancer malignancy has been observed. PDZK1 plays a role in cell growth by enhancing Akt stabilization through increased interaction with Cdc37. PMID: 24869908
  12. The N-terminal tail serves as an intramolecular chaperone, ensuring that CDC37 adopts one of two interconvertible states. This dynamic process influences the interaction of the client binding N-domain and the MC-domains, which are involved in dimerization and HSP90 binding. PMID: 25619116
  13. CDC37 plays a pivotal role in chaperoning protein kinases. It stabilizes kinase clients through a mechanism that does not rely on substantial direct interaction between CDC37 and HSP90, but necessitates HSP90 activity. PMID: 24292678
  14. As a novel Hsp90 inhibitor, FW-04-806 binds to the N-terminal of Hsp90 and inhibits Hsp90/Cdc37 interaction. This disruption leads to the dissociation of Hsp90/Cdc37/client complexes and the degradation of Hsp90 client proteins. PMID: 24927996
  15. CDC37 is a critical HSP90-cofactor for KIT oncogenic expression in gastrointestinal stromal tumors. PMID: 23584476
  16. SGK3 stability and kinase activation are regulated by the Hsp90-Cdc37 chaperone complex. PMID: 24379398
  17. Cdc37 (cell division cycle 37) restricts Hsp90 (heat shock protein 90) motility by interacting with N-terminal and middle domain binding sites. PMID: 23569206
  18. ERK5 interacts with the Hsp90-Cdc37 chaperone in resting cells. Inhibition of Hsp90 or Cdc37 results in ERK5 ubiquitylation and proteasomal degradation. PMID: 23428871
  19. Surface Cdc37, in conjunction with HSP90, plays a crucial role in cancer cell invasion processes. PMID: 22912728
  20. A series of tyrosine phosphorylation events, involving both p50(Cdc37) and Hsp90, are minimally sufficient to provide directionality to the chaperone cycle. PMID: 22727666
  21. Data indicates that a portion of the normal clearance cascade for TDP-43 involves the Cdc37/Hsp90 complex. PMID: 22674575
  22. Cdc37-mediated direct interaction between Hsp90/Cdc37 and an IRE1alpha cytosolic motif is important for maintaining basal IRE1alpha activity and contributes to normal protein homeostasis and unfolded protein response under physiological stimulation. PMID: 22199355
  23. The primary mechanisms by which apigenin eliminates multiple myeloma cells involve targeting the trinity of CK2-Cdc37-Hsp90. PMID: 21871133
  24. The Hsp90-Cdc37 complex acts as an endogenous regulator of noncanonical p38alpha activity. PMID: 20299663
  25. Tnf-induced recruitment and activation of the IKK complex require Cdc37 and Hsp90. PMID: 11864612
  26. Cdc37 plays a role in regulating Hsp90 ATPase activity. PMID: 11916974
  27. CDC37 binds to Akt and HSP90 in the signal transduction pathway within human tumor cells. PMID: 12176997
  28. Results indicate that Cdc37 and heat shock protein 90 bind specifically to the kinase domain of LKB1. PMID: 12489981
  29. Phosphorylation of Cdc37 on Ser13 is crucial for its ability to coordinate Hsp90 nucleotide-mediated conformational switching and kinase binding. PMID: 12930845
  30. Heteromeric complexes containing the molecular chaperones Hsp90 and Cdc37/p50 interact with the kinase domain of LKB1. PMID: 14668798
  31. The interaction of Cdc37 with its client protein kinases requires amino acid residues within a motif that is present in many protein kinases. PMID: 14701845
  32. Hsp90/p50cdc37 is required for mixed-lineage kinase (MLK) 3 signaling. PMID: 15001580
  33. The Hsp90.Cdc37 molecular chaperone module plays a central role in interleukin-1 receptor-associated-kinase-dependent signaling by toll-like receptors. PMID: 15647277
  34. Cdc37 has been found to heterodimerize with heat-shock protein 90 (Hsp90)-associating relative of Cdc37 (Harc) in vitro. PMID: 15850399
  35. Nuclear magnetic resonance study of binding to HSP90. PMID: 16132836
  36. Results suggest that a region of Cdc37, other than the client-binding site, may be responsible for discriminating client protein kinases from others. PMID: 16156789
  37. JAK1/2 are client proteins of Hsp90 alpha and beta; Hsp90 and CDC37 play a critical role in types I and II interferon pathways. PMID: 16280321
  38. The N-terminal glycine-rich loop of protein kinases is essential for physically associating with Cdc37. PMID: 16611982
  39. Data showcases the expression and purification of an Hsp90-Cdc37-Cdk4 complex, defining its stoichiometry, and determining its 3D structure through single-particle electron microscopy. PMID: 16949366
  40. These observations support the hypothesis that a specific coordination exists between the activation of the cytosolic Ah receptor and the c-Src- and cdc37-containing HSP90 complex. PMID: 17223712
  41. The current data identifies Hsp90-Cdc37 as a transiently acting essential regulatory component of IKK signaling. PMID: 17728246
  42. Pink1 is identified as a novel Cdc37/Hsp90 client kinase. PMID: 18003639
  43. Cdc37 is essential for maintaining prostate tumor cell growth and may represent a novel target in the search for multitargeted therapies. PMID: 18089825
  44. These data reveal a cyclic regulatory mechanism for Cdc37, where its constitutive phosphorylation is reversed by targeted dephosphorylation in Hsp90 complexes. PMID: 18922470
  45. CDC37, in collaboration with HSP90, plays an essential role in maintaining oncogenic protein kinase clients including ERBB2, CRAF, CDK4, CDK6, and phosphorylated AKT. PMID: 18931700
  46. The human Cdc37.Hsp90 complex has been studied using heteronuclear NMR spectroscopy. PMID: 19073599
  47. The C-terminal tail and determinants within the alphaE-helix of the catalytic domain allow the chaperones Hsp90 and Cdc37 to bind newly synthesized PKC beta II. This binding is a necessary step in the processing of PKC through phosphorylation. PMID: 19091746
  48. Celastrol may represent a novel class of Hsp90 inhibitor by modifying Hsp90's C terminus to allosterically regulate its chaperone activity and disrupt the Hsp90-Cdc37 complex. PMID: 19858214

Show More

Hide All

Database Links

HGNC: 1735

OMIM: 605065

KEGG: hsa:11140

STRING: 9606.ENSP00000222005

UniGene: Hs.160958

Protein Families
CDC37 family
Subcellular Location
Cytoplasm.

Q&A

What is the biological significance of CDC37 phosphorylation at S13?

CDC37 phosphorylation at serine 13 plays a crucial role in promoting the formation of stable HSP90-CDC37-kinase ternary complexes, which are essential for proper kinase folding and activation. Importantly, S13 phosphorylation does not affect the boundaries of CDC37's predicted secondary structure elements or its ability to form binary CDC37-kinase complexes. Instead, this post-translational modification appears to specifically stabilize the interaction with HSP90, functioning at a later stage in the chaperone cycle after initial kinase recognition . The phosphorylation event does not alter the mode of kinase recognition or binding affinity between CDC37 and kinases, but rather regulates the stability of the ternary complex formation .

What detection methods are compatible with the Phospho-CDC37 (S13) antibody?

The Phospho-CDC37 (S13) Recombinant Monoclonal Antibody has been validated for dot blots and Western blot (WB) analyses . For Western blot applications, the antibody has been tested at a 1/10000 dilution and successfully detects the phosphorylated form of CDC37 at the predicted band size of 44 kDa. Researchers should note that the antibody has been specifically tested with human, mouse, and rat samples, showing consistent results across these species . When conducting Western blot experiments, the antibody can be paired with appropriate secondary antibodies such as Goat Anti-Rabbit IgG H&L (HRP) at a 1/20000 dilution .

How is specificity for phosphorylated S13 validated in experimental settings?

Specificity validation for the Phospho-CDC37 (S13) antibody typically involves phosphatase treatment controls. In Western blot experiments, researchers compare samples with and without phosphatase treatment to confirm specificity for the phosphorylated form of the protein. As demonstrated in validation data, the antibody shows strong reactivity with phosphorylated CDC37 in untreated lysates, while the signal disappears or significantly decreases in phosphatase-treated samples . Additional validation involves using Calyculin A, a phosphatase inhibitor that enhances S13 phosphorylation levels, resulting in stronger signal detection in treated versus untreated samples .

How does S13 phosphorylation impact the structural dynamics of CDC37 in complex formation?

S13 phosphorylation of CDC37 does not alter the protein's mode of kinase recognition or binding affinity. NMR spectroscopy analysis reveals that phosphorylated and non-phosphorylated forms of CDC37 produce identical chemical shift perturbation patterns when interacting with client kinases, such as bRaf . Both forms exhibit the same thermodynamic signatures of binding and identical affinities for client kinases. The current model suggests that S13 phosphorylation primarily functions at later stages of the chaperone cycle, specifically in stabilizing ternary complexes with HSP90 rather than affecting initial kinase recognition . The phosphorylation event appears to act as a regulatory switch that promotes progression through the chaperone cycle after the initial CDC37-kinase binding has occurred.

What are the key experimental considerations when comparing phosphorylated versus non-phosphorylated CDC37 in kinase recognition studies?

When designing experiments to compare phosphorylated versus non-phosphorylated CDC37 in kinase recognition studies, researchers should consider the following methodological approaches:

  • Phosphomimetic mutations: Use S13D or S13E mutations to mimic constitutive phosphorylation and S13A to prevent phosphorylation

  • Controlled phosphorylation: Employ CK2 kinase for in vitro phosphorylation of recombinant CDC37

  • Biochemical validation: Confirm phosphorylation status using the phospho-specific antibody alongside mass spectrometry

  • Binding studies: Implement multiple techniques (ITC, SPR, NMR) to measure binding constants

  • Functional assays: Design experiments that specifically distinguish between binary (CDC37-kinase) and ternary (HSP90-CDC37-kinase) complex formation

Analysis should account for the fact that S13 phosphorylation impacts ternary complex stability without altering binary CDC37-kinase complex formation . Additionally, the S13A mutation affects client interaction and maturation primarily at later stages involving HSP90, with minimal impact on binary complex formation .

How can the Phospho-CDC37 (S13) antibody be used to investigate the role of CDC37 in oncogenic kinase stabilization?

The Phospho-CDC37 (S13) antibody provides a valuable tool for investigating CDC37's involvement in oncogenic kinase stabilization through several methodological approaches:

  • Comparative analysis of phosphorylation levels: Quantify S13 phosphorylation across normal versus cancer cell lines or tissues to establish correlation with oncogenic transformation

  • Kinase-specific interactions: Perform co-immunoprecipitation experiments using the phospho-specific antibody to isolate and identify associated kinases in various cancer models

  • Oncogenic versus wild-type kinase comparisons: As demonstrated with bRaf versus bRaf V600E, CDC37 shows stronger interaction with oncogenic variants ; the phospho-specific antibody can help quantify these differential interactions

  • Pharmacological intervention studies: Monitor changes in S13 phosphorylation following treatment with HSP90 inhibitors, kinase inhibitors, or phosphatase modulators

Research data indicates that oncogenic kinases such as bRaf V600E show significantly stronger interaction with CDC37 compared to their wild-type counterparts . This suggests that phosphorylated CDC37 may play a more critical role in stabilizing oncogenic kinases, making the Phospho-CDC37 (S13) antibody particularly valuable for cancer research applications.

What are the optimal sample preparation protocols for detecting Phospho-CDC37 (S13) in different experimental systems?

For optimal detection of Phospho-CDC37 (S13) in various experimental systems, researchers should implement the following sample preparation protocols:

Cell Culture Samples:

  • Treat cells with phosphatase inhibitors (50nM Calyculin A for 3 hours) to preserve phosphorylation status

  • Harvest cells in buffer containing phosphatase inhibitor cocktails

  • Lyse cells using buffers containing detergents suitable for membrane disruption (RIPA or NP-40)

  • Centrifuge lysates at 14,000g for 15 minutes at 4°C to remove cellular debris

  • Quantify protein concentration using Bradford or BCA assays

  • Load 15-20 μg of total protein per lane for Western blot analysis

Tissue Samples:

  • Snap-freeze tissues immediately after collection

  • Homogenize in cold lysis buffer containing phosphatase inhibitors

  • Clarify lysates by centrifugation

  • Process as with cell culture samples

For validation purposes, researchers should prepare parallel samples treated with phosphatase to confirm antibody specificity for the phosphorylated form of CDC37 .

What experimental controls are essential when using the Phospho-CDC37 (S13) antibody in client kinase interaction studies?

When utilizing the Phospho-CDC37 (S13) antibody in client kinase interaction studies, the following controls are essential for data validation:

Control TypePurposeImplementation Method
Phosphatase treatmentConfirm phospho-specificityTreat duplicate samples with lambda phosphatase prior to Western blotting
Total CDC37 detectionNormalize phosphorylation levelsUse a separate antibody recognizing CDC37 regardless of phosphorylation status
Phosphorylation enhancementPositive controlTreat cells with Calyculin A (50nM, 3 hours) to increase S13 phosphorylation
Knockout/knockdown validationAntibody specificityInclude CDC37-depleted samples to confirm signal specificity
S13A mutantNegative controlExpress CDC37 S13A mutant that cannot be phosphorylated at this site
Cross-reactivity assessmentPrevent false positivesInclude samples containing proteins with similar phosphorylation motifs

Using these controls helps ensure reliable interpretation of results by distinguishing specific phospho-CDC37 (S13) signals from potential artifacts or non-specific binding.

How can Phospho-CDC37 (S13) antibody be used in multiplexed detection systems with other chaperone complex components?

For multiplexed detection of Phospho-CDC37 (S13) alongside other chaperone complex components, researchers can employ several methodological approaches:

  • Sequential immunoblotting: Strip and reprobe membranes with antibodies against HSP90, client kinases, and other co-chaperones, ensuring appropriate molecular weight separation

  • Fluorescent multiplex Western blotting: Utilize primary antibodies from different species and corresponding fluorescently-labeled secondary antibodies with distinct excitation/emission profiles

  • Co-immunoprecipitation followed by immunoblotting:

    • Immunoprecipitate with Phospho-CDC37 (S13) antibody

    • Probe precipitates for HSP90, client kinases, and other components

    • Compare with reciprocal immunoprecipitations using antibodies against complex components

  • Multiplex immunofluorescence microscopy:

    • Use differently labeled secondary antibodies against primaries for Phospho-CDC37 (S13), HSP90, and client kinases

    • Analyze co-localization patterns using confocal microscopy

    • Quantify colocalization coefficients to assess complex formation

  • Proximity ligation assay (PLA):

    • Combine Phospho-CDC37 (S13) antibody with antibodies against HSP90 or kinases

    • Generate fluorescent signals only when proteins are in close proximity (<40nm)

    • Quantify interaction events in situ

These multiplexed approaches allow researchers to simultaneously examine the phosphorylation status of CDC37 and its association with other components of the chaperone machinery in various experimental contexts.

How should researchers interpret changes in CDC37 S13 phosphorylation levels in response to different cellular stresses?

Interpreting changes in CDC37 S13 phosphorylation in response to cellular stresses requires consideration of multiple factors. CDC37 phosphorylation at S13 promotes the formation of stable HSP90-CDC37-kinase ternary complexes essential for kinase maturation and function . Therefore, changes in phosphorylation levels may indicate adaptive responses in the cell's protein quality control system.

When analyzing phosphorylation changes, researchers should:

Increases in S13 phosphorylation often indicate enhanced demand for kinase stabilization, while decreased phosphorylation may suggest reduced chaperone capacity or altered regulatory mechanisms in the stress response.

What are common technical challenges when detecting Phospho-CDC37 (S13) and their solutions?

Researchers commonly encounter several technical challenges when working with Phospho-CDC37 (S13) antibody. The following table outlines these challenges and provides methodological solutions:

ChallengePossible CausesSolutions
Weak or absent signalDephosphorylation during sample preparationInclude phosphatase inhibitors (Calyculin A at 50nM) in all buffers; maintain samples at 4°C
Multiple bandsCross-reactivity with similar phosphomotifsIncrease antibody dilution (1/10000 or higher); validate with phosphatase treatment controls
High backgroundNon-specific bindingUse longer blocking times (2+ hours); increase concentration of blocking reagent; optimize antibody dilution
Inconsistent results between replicatesVariable phosphorylation levelsStandardize cell culture conditions; ensure consistent treatment times; validate phosphorylation with phosphatase controls
Loss of signal during stripping/reprobingEpitope sensitivityAvoid harsh stripping conditions; consider using parallel blots instead of stripping
Species cross-reactivity issuesEpitope sequence variationValidate antibody in your specific species; consider using synthetic phosphopeptides as controls

When troubleshooting, researchers should remember that the Phospho-CDC37 (S13) antibody has been validated specifically for dot blots and Western blot applications at defined concentrations (1/10000 dilution) .

How does Phospho-CDC37 (S13) status correlate with the stability and activity of client kinases across different experimental models?

The correlation between Phospho-CDC37 (S13) status and client kinase stability/activity varies across experimental models, reflecting the complex regulation of the HSP90-CDC37-kinase chaperone system. Research findings indicate:

  • Cancer models: Enhanced S13 phosphorylation often correlates with increased stability of oncogenic kinases like bRaf V600E, which shows significantly stronger interaction with CDC37 compared to wild-type bRaf . This suggests that phosphorylated CDC37 may preferentially stabilize oncogenic kinase variants.

  • Cell type specificity: Different cell types show variable dependency on CDC37 phosphorylation for kinase stability, likely reflecting tissue-specific chaperone networks.

  • Kinase-specific effects: The impact of S13 phosphorylation appears to be more pronounced for certain kinases. While S13 phosphorylation does not directly affect the mode of kinase recognition or binding affinity of CDC37 to kinases like bRaf , it enhances stability of ternary complexes involving HSP90.

  • Temporal dynamics: The relationship between phosphorylation and kinase activity shows time-dependent patterns during cellular processes like differentiation and stress response.

  • Pharmacological interventions: HSP90 inhibitors can disrupt the stability provided by phosphorylated CDC37, leading to degradation of client kinases, suggesting that targeting this phosphorylation could be a therapeutic strategy.

When designing experiments to investigate these correlations, researchers should employ multiple approaches, including phosphorylation-site mutants (S13A, S13D), pharmacological modulators of CK2 activity, and client kinase activity/stability assays .

How can researchers utilize the Phospho-CDC37 (S13) antibody to investigate the conformational changes in CDC37 upon phosphorylation?

Investigating conformational changes in CDC37 upon S13 phosphorylation requires sophisticated biophysical approaches combined with antibody-based detection. Researchers can employ the following methodological strategies:

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Compare exchange patterns between phosphorylated and non-phosphorylated CDC37

    • Use the Phospho-CDC37 (S13) antibody to validate phosphorylation status of samples

    • Correlate regions of altered solvent accessibility with functional domains

  • Limited proteolysis coupled with Western blotting:

    • Subject phosphorylated and non-phosphorylated CDC37 to controlled proteolytic digestion

    • Analyze fragment patterns using the Phospho-CDC37 (S13) antibody and total CDC37 antibodies

    • Identify regions with altered protease sensitivity indicating conformational differences

  • Cross-linking mass spectrometry:

    • Apply chemical cross-linkers to capture the three-dimensional structure

    • Identify crosslinked peptides by mass spectrometry

    • Compare crosslinking patterns between phosphorylated and non-phosphorylated forms

  • Single-molecule FRET combined with immunovalidation:

    • Engineer CDC37 with fluorophore pairs at strategic positions

    • Measure FRET efficiency changes upon phosphorylation

    • Validate phosphorylation status using the Phospho-CDC37 (S13) antibody

Current evidence indicates that S13 phosphorylation does not significantly alter the predicted secondary structure elements of N-Cdc37 , suggesting that its effects may involve more subtle conformational changes or alterations in protein-protein interaction surfaces that facilitate stable ternary complex formation with HSP90 and client kinases.

What approaches can be used to investigate the differential effects of S13 phosphorylation on various client kinase interactions?

To investigate how S13 phosphorylation differentially affects interactions with various client kinases, researchers can implement the following methodological approaches:

  • Comparative binding assays:

    • Perform quantitative pull-down experiments with phosphorylated versus non-phosphorylated CDC37

    • Compare binding affinities across a panel of client and non-client kinases

    • Use the Phospho-CDC37 (S13) antibody to confirm phosphorylation status

  • Kinase specificity profiling:

    • Develop protein microarrays with diverse kinases

    • Probe with phosphorylated versus non-phosphorylated CDC37

    • Quantify binding differences using detection systems including the Phospho-CDC37 (S13) antibody

  • Structural characterization of complexes:

    • Use cryo-EM to visualize CDC37-kinase complexes with and without S13 phosphorylation

    • Perform hydrogen-deuterium exchange mass spectrometry on complexes with different kinases

    • Identify kinase-specific interaction interfaces affected by phosphorylation

  • Client kinase stability assays:

    • Express CDC37 wild-type, S13A, and S13D/E mutants in cells

    • Measure half-lives of different client kinases under each condition

    • Correlate with phosphorylation status using the Phospho-CDC37 (S13) antibody

Research indicates that while S13 phosphorylation does not alter binary CDC37-kinase complex formation, it significantly impacts the formation of stable ternary complexes with HSP90 . The effect appears to be more pronounced with certain kinases, particularly oncogenic variants like bRaf V600E, which shows stronger interaction with CDC37 compared to wild-type bRaf .

How can multi-omics approaches incorporating Phospho-CDC37 (S13) detection advance our understanding of chaperone networks in disease models?

Multi-omics approaches integrating Phospho-CDC37 (S13) detection can significantly enhance our understanding of chaperone networks in disease models through systematic analysis of multiple biological layers:

  • Phosphoproteomics and Interactomics integration:

    • Perform phospho-enrichment coupled with mass spectrometry across disease models

    • Use the Phospho-CDC37 (S13) antibody for validation and quantification

    • Correlate CDC37 S13 phosphorylation with global phosphorylation networks

    • Perform parallel interactome analysis using co-immunoprecipitation with the phospho-specific antibody

    • Construct interaction networks specific to phosphorylated versus non-phosphorylated CDC37

  • Transcriptomics and Proteomics correlation:

    • Compare transcriptional profiles of cells with normal versus altered CDC37 phosphorylation

    • Correlate with protein abundance changes of chaperone components and client kinases

    • Use the Phospho-CDC37 (S13) antibody to stratify samples based on phosphorylation status

  • Functional genomics screens:

    • Perform CRISPR screens to identify genes affecting CDC37 S13 phosphorylation

    • Use the phospho-specific antibody as a readout for screen validation

    • Connect genetic dependencies with CDC37 phosphorylation status in disease models

  • Metabolomics integration:

    • Correlate metabolic signatures with CDC37 phosphorylation status

    • Investigate how metabolic states influence chaperone function via CDC37 modification

    • Examine feedback loops between kinase signaling and metabolism mediated by CDC37

This multi-dimensional analysis can reveal how CDC37 S13 phosphorylation serves as a regulatory node within larger cellular networks, potentially identifying new therapeutic targets for diseases dependent on specific chaperone-client interactions.

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.