Phospho-RAF1 (S259) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Phospho-RAF1 (S259) Recombinant Monoclonal Antibody

The Phospho-RAF1 (S259) Recombinant Monoclonal Antibody is a highly specific tool designed to detect phosphorylation at serine residue 259 (S259) of the RAF1 protein. RAF1 is a critical kinase in the RAS-RAF-MEK-ERK signaling pathway, regulating cell proliferation, differentiation, and survival . Phosphorylation at S259 is a key regulatory event that modulates RAF1 activity, membrane localization, and interactions with downstream targets . This antibody is engineered to bind exclusively to the phosphorylated form of RAF1, enabling precise analysis of signaling dynamics in cancer, apoptosis, and cellular stress responses .

Production and Specificity

This recombinant monoclonal antibody is produced using advanced technologies to ensure high specificity:

  • Immunogen: A synthetic phosphorylated peptide corresponding to residues surrounding S259 of human RAF1 .

  • Host: Generated in rabbits, leveraging their robust immune response to phosphorylated epitopes .

  • Purification: Affinity chromatography and peptide affinity purification to isolate the antibody from host cell supernatant .

Key Features:

ParameterDetails
IsotypeIgG (monoclonal)
ReactivityHuman (primary), Rat (secondary)
Observed MW73–75 kDa (matches predicted RAF1 size)
ApplicationsWestern blot (WB), Immunohistochemistry (IHC), Immunofluorescence (IF)

Western Blot (WB)

  • Dilution: 1:500–1:5,000 .

  • Example Protocol:

    • Sample Preparation: Treat HEK293 cells with phorbol esters (e.g., PMA) to activate RAF1 phosphorylation .

    • Detection: Use secondary anti-rabbit HRP-conjugated antibodies (e.g., ab97051) at 1:20,000 dilution .

    • Controls: Incubate lysates with phosphatases to confirm signal specificity .

Immunohistochemistry (IHC) and Immunofluorescence (IF)

  • Dilution: IHC (1:50–1:200), IF (1:20–1:200) .

  • Utility: Localizes phosphorylated RAF1 in tissues or cells, aiding studies on subcellular distribution (e.g., membrane vs. cytoplasmic signaling) .

Role of S259 Phosphorylation in RAF1 Activity

Phosphorylation at S259 regulates RAF1’s ability to interact with downstream targets:

  • Membrane Localization: S259 phosphorylation impairs RAF1’s association with the plasma membrane, modulating its activation by Ras .

  • 14-3-3 Binding: While S621 phosphorylation is dispensable, 14-3-3 proteins bind to RAF1’s CR3 domain to stabilize its activity .

  • Cancer Relevance: Dysregulation of S259 phosphorylation is implicated in oncogenic signaling, particularly in cancers with RAS/RAF pathway hyperactivation .

Mechanistic Implications

  • Cross-Talk with Other Pathways: S259 phosphorylation may influence RAF1’s interaction with adenylyl cyclases, ROCK2, or BAD/Bcl2 .

  • Therapeutic Potential: Inhibitors targeting S259 phosphorylation could disrupt oncogenic RAF1 signaling, though clinical validation is pending .

Product Specs

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

The phospho-RAF1 (S259) recombinant monoclonal antibody is produced through a multi-step process. First, rabbit antibody genes are sourced from animals immunized with a synthetic peptide representing human RAF1 phosphorylated at serine 259 (S259). These genes are then cloned into expression vectors and transfected into mammalian host cells. Following cell culture and antibody secretion, the phospho-RAF1 (S259) antibody is purified using affinity chromatography. Rigorous functional validation is performed using ELISA, Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (IF) assays to confirm its specific binding to human RAF1 phosphorylated at S259.

Phosphorylation of RAF1 at S259 is a crucial regulatory event within the mitogen-activated protein kinase (MAPK) signaling pathway. This modification plays a significant role in controlling cellular processes such as proliferation, differentiation, and survival. Dysregulation of S259 phosphorylation is implicated in the pathogenesis of cancer and other diseases characterized by aberrant signaling pathways.

Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
c Raf antibody; C-raf antibody; C-Raf proto-oncogene, serine/threonine kinase antibody; CMD1NN antibody; Craf 1 transforming gene antibody; cRaf antibody; Craf1 transforming gene antibody; EC 2.7.11.1 antibody; kinase Raf1 antibody; Murine sarcoma 3611 oncogene 1 antibody; NS5 antibody; Oncogene MIL antibody; Oncogene RAF1 antibody; OTTHUMP00000160218 antibody; OTTHUMP00000207813 antibody; OTTHUMP00000209389 antibody; Protein kinase raf 1 antibody; Proto-oncogene c-RAF antibody; Raf 1 antibody; Raf 1 proto oncogene serine/threonine kinase antibody; RAF antibody; Raf proto oncogene serine/threonine protein kinase antibody; RAF proto-oncogene serine/threonine-protein kinase antibody; RAF-1 antibody; RAF1 antibody; RAF1_HUMAN antibody; Similar to murine leukemia viral (V-raf-1) oncogene homolog 1 antibody; TRANSFORMING REPLICATION-DEFECTIVE MURINE RETROVIRUS 3611-MSV antibody; v raf 1 murine leukemia viral oncogene homolog 1 antibody; v-raf murine sarcoma viral oncogene homolog 1 antibody; v-raf-1 murine leukemia viral oncogene-like protein 1 antibody; vraf1 murine leukemia viral oncogene homolog 1 antibody
Target Names
Uniprot No.

Target Background

Function

RAF1 is a serine/threonine-protein kinase that acts as a critical regulatory link between membrane-bound Ras GTPases and the MAPK/ERK cascade. This function serves as a molecular switch controlling various cellular fate decisions, including proliferation, differentiation, apoptosis, survival, and oncogenic transformation. RAF1 activation initiates a MAPK cascade involving sequential phosphorylation of MAP2K1/MEK1, MAP2K2/MEK2, MAPK3/ERK1, and MAPK1/ERK2. Phosphorylated RAF1 (at Ser-338 and Ser-339, mediated by PAK1) phosphorylates BAD, leading to its inactivation. It also phosphorylates and activates adenylyl cyclases ADCY2, ADCY5, and ADCY6; inhibits the phosphatase activity of PPP1R12A; and phosphorylates TNNT2. Furthermore, RAF1 can modulate NF-κB activation, inhibit motility-related signal transducers (ROCK2), apoptosis-related kinases (MAP3K5/ASK1 and STK3/MST2), and factors involved in proliferation and angiogenesis (RB1). RAF1 can protect against apoptosis by mitochondrial translocation, binding to BCL2 and displacing BAD. It also regulates Rho signaling and migration, and is essential for normal wound healing. In epithelial cells, RAF1 contributes to oncogenic transformation by repressing the tight junction protein occludin (OCLN) through upregulation of the transcriptional repressor SNAI2/SLUG. Finally, RAF1 can restrict caspase activation in response to specific stimuli, such as Fas stimulation, pathogen-mediated macrophage apoptosis, and erythroid differentiation.

Gene References Into Functions
  1. Functional assessment of RAF1 and RIT1 variants of uncertain significance (VUSs); unclear significance of A2ML1 VUSs. PMID: 29402968
  2. Familial Noonan syndrome case due to a germline RAF1 p.S427G substitution; lack of association with increased tumor risk. PMID: 30204961
  3. RAF1 acts as a negative regulator of hepatocarcinogenesis. PMID: 28000790
  4. Inherited RAF1-associated Noonan syndrome case presenting with antenatal skull abnormalities, subdural hematomas, delayed myelination, and polymicrogyria. PMID: 27753652
  5. Raf-1 as a potential prognostic factor and therapeutic target in non-small cell lung cancer (NSCLC). PMID: 29484414
  6. Regulation of RAF1 binding to SPRY4 by miR-1908 in gliomas. PMID: 29048686
  7. Association of high RAF1 expression with malignant melanoma. PMID: 28677804
  8. Two premature neonates with biventricular hypertrophy and RAF1 variants in the CR2 domain. PMID: 28777121
  9. CNK1 as a regulator of RAF and AKT signaling and cell fate decisions. PMID: 27901111
  10. CRAF as an alternative oncogene in BRAF/NRAS/GNAQ/GNA11 wild-type melanomas. PMID: 27273450
  11. miR-125a as a tumor suppressor regulating sirtuin-7, MMP-11, and c-Raf. PMID: 28445974
  12. ciRS-7 overexpression inhibits miR-7, activating EGFR and RAF1 oncogenes. PMID: 28174233
  13. miR-497 as a potential tumor suppressor and diagnostic marker in gastric cancer by targeting Raf-1. PMID: 28586056
  14. RAF1's potential role in hepatocellular carcinoma survival and sorafenib adjuvant therapy. PMID: 26981887
  15. Mutational activation of Kit, Ras/Raf/Erk, and Akt pathways as therapeutic targets. PMID: 27391150
  16. DCP antagonizes Sorafenib's inhibitory effects on HCC via Raf/MEK/ERK and PI3K/Akt/mTOR pathways. PMID: 27167344
  17. DiRas3 binds to KSR1 independently of its interaction with activated Ras and RAF. PMID: 27368419
  18. RhoA/ROCK and Raf-1/CK2 pathways mediate TNF-α-induced endothelial cytotoxicity via vimentin cytoskeleton regulation. PMID: 28743511
  19. Raf-1 kinase feedback regulation abnormality enhances antiapoptotic function; Raf-1 as a potential therapeutic target to increase chemotherapy/radiotherapy sensitivity. PMID: 27841865
  20. RAF1's critical role in maintaining the transformed phenotype of colorectal cancer (CRC) cells, including KRAS-mutated cells. PMID: 27670374
  21. Hsp90's role in CRAF maturation and activation as a limiting factor in maintaining CRAF kinase function. PMID: 27703006
  22. Oncogenic NFIA:RAF1 fusion activation of the MAPK pathway in pilocytic astrocytoma. PMID: 27810072
  23. IGF2BP2's role in colorectal carcinogenesis by interfering with Raf-1 degradation by miR-195. PMID: 27153315
  24. miR-125b overexpression reduces RAF1 expression and promotes macrophage apoptosis. PMID: 27363278
  25. Griffipavixanthone (GPX) as a B-RAF and C-RAF inhibitor in esophageal cancer cells. PMID: 26646323
  26. Association of Raf-1 upregulation with triple-negative breast cancer. PMID: 26513016
  27. Molecular basis for C-Raf C-terminal phosphopeptide interaction with 14-3-3ζ protein. PMID: 26295714
  28. CD166 regulates MCAM via PI3K/AKT and c-Raf/MEK/ERK signaling and inhibition of betaTrCP and Smurf1 ubiquitin E3 ligases. PMID: 26004137
  29. Interrelated kinase module involving c-Raf/PI3K/Lyn and Fgr in retinoic acid-induced maturation or rescue of RA-resistant leukemia cells. PMID: 25817574
  30. Role of aberrant Ras/MAPK pathway activation in pulmonary vascular disease in Noonan syndrome with specific RAF1 mutations. PMID: 25706034
  31. Raf-1 as a potential prognostic biomarker in chordoma. PMID: 25755752
  32. Structural effects of RasQ61L mutation on Raf-Ras binding domain flexibility. PMID: 25684575
  33. pDAPK(S308) as a potential predictive biomarker for Raf inhibitor combination therapy. PMID: 26100670
  34. DJ-1's role in stimulating c-Raf self-phosphorylation and subsequent MEK and ERK1/2 phosphorylation. PMID: 26048984
  35. Truncated RAF1 and BRAF proteins conferring resistance to RTK-targeted therapy in cancer cells. PMID: 25473895
  36. miR-455-RAF1 as a potential therapeutic target for colorectal carcinoma. PMID: 25355599
  37. Identification of 18 kinase and kinase-related genes (including RAF1) that can substitute for EGFR in EGFR-dependent cells. PMID: 25512530
  38. Frequent aberrant expression of A-, B-, and C-RAF, and COT in papillary thyroid cancer (PTC); correlation between COT overexpression and PTC recurrence. PMID: 25674762
  39. NMR analysis of human Raf1 N-terminus (hRaf11-147aa) binding to human RKIP (hRKIP). PMID: 24863296
  40. c-Raf among anti-apoptotic Bcl-2 family members. PMID: 24969872
  41. miR-7-5p as a tumor suppressor regulating glioblastoma microvascular endothelial cell proliferation by targeting RAF1. PMID: 25027403
  42. CAV-1 disruption of BRaf/CRaf heterodimer and inhibition of MAPK pathway during dasatinib treatment. PMID: 24486585
  43. Regulation of RAF-1 ubiquitination and levels by Shoc2 and HUWE1. PMID: 25022756
  44. Raf-1/JNK/p53/p21 pathway involvement in apoptosis; NFκB1's potential role in inhibiting apoptosis. PMID: 22282237
  45. Higher RAF1 mRNA expression and AKT/ERK activation conferring vinorelbine resistance in non-small cell lung cancer. PMID: 24427333
  46. Analysis of RAF1 mutations in childhood-onset dilated cardiomyopathy patients. PMID: 24777450
  47. miR-195 expression or Raf-1 knockdown reducing tumor cell survival. PMID: 23760062
  48. Potential role of SRC, RAF1, and PTK2B genes in neurotransmission and CNS signaling. PMID: 24108181
  49. Identification of C-RAF mutations causing biochemical and pharmacologic resistance in melanoma cell lines. PMID: 23737487
  50. ARAF stabilization of BRAF:CRAF complexes and regulation of cell signaling in response to RAF inhibitors. PMID: 22926515
Database Links

HGNC: 9829

OMIM: 164760

KEGG: hsa:5894

STRING: 9606.ENSP00000251849

UniGene: Hs.159130

Involvement In Disease
Noonan syndrome 5 (NS5); LEOPARD syndrome 2 (LPRD2); Cardiomyopathy, dilated 1NN (CMD1NN)
Protein Families
Protein kinase superfamily, TKL Ser/Thr protein kinase family, RAF subfamily
Subcellular Location
Cytoplasm. Cell membrane. Mitochondrion. Nucleus. Note=Colocalizes with RGS14 and BRAF in both the cytoplasm and membranes. Phosphorylation at Ser-259 impairs its membrane accumulation. Recruited to the cell membrane by the active Ras protein. Phosphorylation at Ser-338 and Ser-339 by PAK1 is required for its mitochondrial localization. Retinoic acid-induced Ser-621 phosphorylated form of RAF1 is predominantly localized at the nucleus.
Tissue Specificity
In skeletal muscle, isoform 1 is more abundant than isoform 2.

Q&A

What is RAF1 and what role does it play in cellular signaling pathways?

RAF1 is a serine/threonine-protein kinase that functions as a critical regulatory link between membrane-associated Ras GTPases and the MAPK/ERK cascade. This regulatory link serves as a molecular switch determining various cell fate decisions including proliferation, differentiation, apoptosis, survival, and oncogenic transformation. RAF1 activation initiates a mitogen-activated protein kinase (MAPK) cascade comprising sequential phosphorylation of the dual-specific MAPK kinases (MAP2K1/MEK1 and MAP2K2/MEK2) and the extracellular signal-regulated kinases (MAPK3/ERK1 and MAPK1/ERK2) .

What is the biological significance of S259 phosphorylation in RAF1 regulation?

Phosphorylation of RAF1 at serine 259 is a crucial regulatory event in cell signaling pathways and cancer development. This phosphorylation event negatively regulates RAF1 activity and serves as part of the control mechanism for cell growth and transformation. When S259 is phosphorylated (typically by kinases such as AKT or PKA), RAF1 activity is suppressed, whereas dephosphorylation by protein phosphatase 2A (PP2A) has been reported to be an essential step in the RAF1 activation mechanism . Understanding the phosphorylation status of RAF1 can provide important insights into the mechanisms underlying oncogenesis and potentially offer therapeutic targets for cancer treatment .

How does the RAF1 (S259) phosphorylation status affect downstream MAPK pathway activation?

The phosphorylation status of RAF1 at S259 directly impacts MAPK pathway activation. When S259 is phosphorylated, RAF1 remains in an inactive conformation, preventing downstream signal transduction. Experimental evidence shows that mutation of S259 to alanine (S259A), which prevents phosphorylation at this site, leads to elevated ERK phosphorylation without additional stimulation . This confirms that dephosphorylation of S259 is a critical step in allowing RAF1 to activate the MAPK pathway. The regulation of S259 phosphorylation thus serves as a control point for modulating the intensity and duration of MAPK signaling in response to external stimuli .

What is the relationship between 14-3-3 protein binding and S259 phosphorylation in RAF1 regulation?

Recent research has revealed that 14-3-3 binding to RAF1, rather than S259 phosphorylation itself, is the essential regulatory mechanism for RAF1 activity. When 14-3-3 binding to conserved region 2 (CR2) of RAF1 is disrupted, RAF1 basal kinase activity becomes elevated and can be further activated. The 14-3-3 proteins bind to RAF1 at phosphorylated serine residues, with S259 serving as one of the primary binding sites. This interaction maintains RAF1 in an inactive conformation until appropriate cellular signals trigger its activation .

Experiments have shown that mutations that prevent 14-3-3 binding to the CR2 region result in increased basal RAF1 activity, demonstrating that 14-3-3 proteins act as negative regulators of RAF1 through this interaction. Furthermore, it has been shown that 14-3-3 binding to CR3 protects S621 from dephosphorylation, which is necessary to maintain RAF1 activity, and also protects active RAF1 from PP1- and PP2A-mediated inactivation .

How does the ERK-phosphorylated RAF1 pool differ from the total RAF1 population in terms of kinase activity?

Research has demonstrated that the ERK-phosphorylated RAF1 pool exhibits approximately 4 times higher specific kinase activity than the total RAF1 population. Importantly, the phosphopeptide composition of this highly active pool is similar to that of the general RAF1 population. This suggests that the preexisting, phosphorylated RAF1, which represents the activatable RAF1 pool, is the specific RAF1 subpopulation targeted by ERK .

Functionally, ERK-1 expression sustains RAF1 activation in a manner dependent on RAF1 phosphorylation on specific identified sites. When these sites are mutated (S289/296/301A substitution), there is a marked decrease in the in vivo activity of RAF1 S259A. This provides evidence for a positive feedback mechanism in RAF1 regulation, where ERK phosphorylation of RAF1 enhances and sustains its activation state .

What are the implications of simultaneous versus separate phosphorylation of S259 and S621 in RAF1?

An important unresolved question in RAF1 research is whether phosphorylation at S259 and S621 occurs simultaneously on the same RAF1 protein or whether they represent two separate RAF1 populations. This question has significant implications for understanding RAF1 regulation mechanisms .

Current evidence suggests that while S259 phosphorylation is associated with inactive RAF1 and 14-3-3-mediated suppression, S621 phosphorylation provides a positive binding point for 14-3-3 that is critical for RAF1 kinase activity. The relationship between these two phosphorylation events remains complex, with some researchers proposing that they may represent different functional states of RAF1 populations within the cell .

Phosphorylation SiteEffect on RAF1 Activity14-3-3 BindingRegulatory Mechanism
S259InhibitoryYesMaintains inactive conformation
S621ActivatingYesCritical for kinase activity

What are the optimal applications and conditions for using Phospho-RAF1 (S259) antibodies in experimental workflows?

Phospho-RAF1 (S259) antibodies are versatile tools that can be employed in multiple experimental applications. Based on validated protocols, these antibodies are suitable for Western blot (WB), immunohistochemistry (IHC-P), immunocytochemistry/immunofluorescence (ICC/IF), ELISA, and dot blot applications .

For Western blot applications, the recommended dilution range is typically 1:500-1:5000, with optimal results generally achieved around 1:1000 . When using these antibodies for immunohistochemistry, a dilution range of 1:50-1:200 is recommended . For immunofluorescence applications, researchers should consider dilutions between 1:20-1:200 .

The antibodies demonstrate strong reactivity with human samples and, depending on the specific clone, may also react with mouse and rat samples . When designing experiments, it's important to confirm the species reactivity of the specific antibody being used.

How can researchers effectively validate the specificity of Phospho-RAF1 (S259) antibodies in their experimental systems?

Validating antibody specificity is crucial for ensuring reliable experimental results. For Phospho-RAF1 (S259) antibodies, several validation approaches can be employed:

  • Phosphatase treatment: Treating lysates with lambda phosphatase should eliminate the signal if the antibody is truly phospho-specific.

  • Phospho-null mutants: Using S259A mutant RAF1 as a negative control can confirm antibody specificity. As demonstrated in published research, the pS259 antibody binds to wild-type RAF1 and A621RAF1 but not to A259RAF1, confirming its specificity for phosphorylation at S259 .

  • Stimulation/inhibition experiments: Treating cells with stimuli known to affect S259 phosphorylation (such as AKT or PKA inhibitors/activators) should result in predictable changes in antibody signal intensity.

  • Peptide competition assays: Pre-incubating the antibody with the phosphorylated peptide used as the immunogen should block specific binding.

What considerations should be made when selecting between monoclonal and polyclonal Phospho-RAF1 (S259) antibodies?

When choosing between monoclonal and polyclonal Phospho-RAF1 (S259) antibodies, researchers should consider several factors:

Monoclonal antibodies offer advantages including:

  • Higher specificity for the exact phospho-epitope

  • Greater batch-to-batch consistency

  • Reduced background in applications like IHC and IF

  • Superior performance in quantitative applications

The recombinant monoclonal antibodies, such as those derived from rabbit antibody genes obtained from animals immunized with synthetic phosphorylated peptides around S259 of human RAF1, offer particularly high specificity and sensitivity for detecting this critical protein modification .

Polyclonal antibodies may offer:

For most precise research applications focusing specifically on S259 phosphorylation status, recombinant monoclonal antibodies are generally preferable due to their consistent performance and high specificity for the phosphorylated form of RAF1 at this specific residue.

How can researchers address issues with non-specific binding when using Phospho-RAF1 (S259) antibodies?

Non-specific binding can compromise experimental results when working with phospho-specific antibodies. To minimize these issues when using Phospho-RAF1 (S259) antibodies, researchers should consider the following strategies:

  • Optimize blocking conditions: Test different blocking agents (BSA, non-fat dry milk, commercial blockers) to identify the most effective option for reducing background.

  • Titrate antibody concentration: Determine the minimum effective concentration by testing serial dilutions beyond the manufacturer's recommended range.

  • Include appropriate controls: Always include negative controls (phosphatase-treated samples or S259A mutants) to distinguish between specific and non-specific signals.

  • Optimize incubation conditions: Adjust temperature, duration, and buffer composition for both primary and secondary antibody incubations.

  • Use phosphatase inhibitors: Ensure complete preservation of phosphorylation status by using fresh phosphatase inhibitors in lysis buffers.

  • Consider alternative detection methods: If conventional methods yield high background, consider more sensitive detection systems or alternative applications (e.g., dot blot vs. Western blot).

How should contradictory results between Phospho-RAF1 (S259) antibody detection and functional RAF1 activity be interpreted?

When researchers encounter discrepancies between Phospho-RAF1 (S259) antibody detection and functional RAF1 activity assays, several factors should be considered:

  • 14-3-3 binding status: As demonstrated in the literature, 14-3-3 binding rather than S259 phosphorylation itself may be the critical determinant of RAF1 activity. Therefore, changes in 14-3-3 binding capacity without changes in S259 phosphorylation could explain such discrepancies .

  • Additional regulatory phosphorylation sites: RAF1 activity is regulated by multiple phosphorylation sites beyond S259. Research has identified novel in vivo RAF1 phosphorylation sites targeted by ERK that provide a positive feedback mechanism for RAF1 regulation . Activity may be influenced by these sites even when S259 phosphorylation status remains unchanged.

  • Context-dependent regulation: The cellular context, including the presence of scaffold proteins, other signaling molecules, and subcellular localization, can significantly impact RAF1 activity independently of S259 phosphorylation.

  • Temporal dynamics: The timing of measurements may influence results, as RAF1 regulation involves dynamic phosphorylation/dephosphorylation events and protein interactions.

When interpreting contradictory results, researchers should consider employing multiple complementary techniques to assess both phosphorylation status and functional activity, ideally with appropriate time-course analyses.

What approaches can be used to study the dynamic regulation of RAF1 S259 phosphorylation in living cells?

Studying the dynamic regulation of RAF1 S259 phosphorylation in living cells requires sophisticated approaches beyond standard fixed-cell techniques:

  • FRET-based biosensors: Design or utilize Förster resonance energy transfer biosensors that can report on S259 phosphorylation status in real-time. These may incorporate phospho-binding domains (such as 14-3-3) that interact with phosphorylated S259.

  • Phospho-specific intrabodies: Develop cell-permeable antibody fragments or intrabodies specifically recognizing phospho-S259 RAF1 that can be expressed within cells.

  • Optogenetic approaches: Combine optogenetic control of upstream kinases/phosphatases with real-time readouts of RAF1 activity to study the temporal relationship between S259 phosphorylation and RAF1 function.

  • Live-cell microscopy with rapid fixation: Employ techniques that allow stimulus application followed by rapid fixation at precise timepoints, combined with phospho-S259 immunostaining.

  • Phosphoproteomics with SILAC or TMT labeling: Use quantitative mass spectrometry approaches with stable isotope labeling to track changes in S259 phosphorylation relative to other phosphorylation sites on RAF1 and related proteins.

These approaches can provide valuable insights into the kinetics and spatial regulation of S259 phosphorylation events that are not possible with traditional biochemical techniques.

What are the implications of RAF1 S259 phosphorylation status for cancer therapy development?

The phosphorylation status of RAF1 at S259 has significant implications for cancer therapy development, particularly for treatments targeting the RAS-RAF-MEK-ERK pathway. Understanding the regulatory mechanisms involving S259 phosphorylation can inform several therapeutic strategies:

  • Direct targeting of S259 regulatory mechanisms: Developing compounds that promote or stabilize S259 phosphorylation could potentially inhibit aberrant RAF1 activation in cancer cells. This approach would differ from current RAF inhibitors that target the kinase domain directly.

  • Combination therapy rationales: Knowledge of how S259 phosphorylation relates to resistance mechanisms can inform rational combination therapies. For example, combining drugs that promote S259 phosphorylation with existing RAF or MEK inhibitors might prevent or delay resistance development.

  • Biomarker development: The phosphorylation status of RAF1 at S259 could serve as a biomarker for predicting response to targeted therapies, allowing for more personalized treatment approaches.

  • Novel drug target identification: Understanding the enzymes responsible for S259 phosphorylation and dephosphorylation in different cancer contexts may reveal new druggable targets in the pathway.

Research has shown that phosphorylation of RAF1 at S259 is a critical event in the control of cell growth and transformation, making it a valuable focal point for developing strategies to control dysregulated MAPK signaling in cancer .

How does cross-talk between the PI3K/AKT and MAPK pathways influence RAF1 S259 phosphorylation?

The cross-talk between PI3K/AKT and MAPK pathways in relation to RAF1 S259 phosphorylation represents an important area of investigation. Research has demonstrated several key aspects of this interaction:

  • AKT-mediated RAF1 inhibition: AKT has been shown to directly phosphorylate RAF1 at S259, resulting in RAF1 inhibition. This represents a critical node of cross-talk between these two major signaling pathways .

  • Pathway integration: The ability of AKT to phosphorylate and inhibit RAF1 provides a mechanism for the PI3K pathway to modulate MAPK signaling, allowing for integrated cellular responses to multiple stimuli.

  • Feedback regulation: Changes in MAPK pathway activity can influence PI3K/AKT signaling, creating complex feedback loops that dynamically regulate RAF1 S259 phosphorylation.

  • Therapeutic implications: Understanding this cross-talk has important implications for combination therapies targeting both pathways, as inhibition of one pathway may lead to compensatory activation of the other through changes in RAF1 S259 phosphorylation.

Future research in this area will likely focus on the context-dependent nature of this cross-talk and how it varies across different cell types and disease states.

What role does RAF1 S259 phosphorylation play in non-canonical RAF1 functions beyond MAPK activation?

Beyond its canonical role in MAPK pathway activation, RAF1 has several non-canonical functions that may be regulated by S259 phosphorylation:

  • Anti-apoptotic functions: RAF1 can translocate to the mitochondria where it binds BCL2 and displaces BAD/Bcl2-antagonist of cell death, protecting cells from apoptosis . The influence of S259 phosphorylation on this non-canonical function remains an important area for investigation.

  • Regulation of cell motility: RAF1 can inhibit signal transducers involved in motility (ROCK2) and plays a role in regulating Rho signaling and migration . Understanding how S259 phosphorylation affects these functions could reveal new insights into processes like metastasis.

  • Epithelial-mesenchymal transition: RAF1 plays a role in the oncogenic transformation of epithelial cells via repression of tight junction proteins through transcriptional regulation . The relationship between S259 phosphorylation and this function represents an emerging research area.

  • Regulation of other kinases: RAF1 can inhibit signal transducers involved in apoptosis (MAP3K5/ASK1 and STK3/MST2) and angiogenesis (RB1) . The dependency of these interactions on S259 phosphorylation status requires further investigation.

Understanding how S259 phosphorylation influences these non-canonical functions could reveal new therapeutic opportunities and explain context-dependent effects of RAF pathway inhibition in different tissues and disease states.

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.