Phospho-RAF1 (S43) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Production and Validation

The antibody is produced via recombinant technology, ensuring batch-to-batch consistency and animal-free production . Validation involves rigorous testing for specificity and functionality:

Validation Methods

TechniqueDetailsSource
ELISAConfirms binding affinity to phosphorylated RAF1 peptides
ImmunofluorescenceDemonstrates localization of phosphorylated RAF1 in HeLa cells (e.g., clone 1F7)
Western BlotDetects phosphorylated RAF1 in cell lysates (e.g., HeLa)

Applications in Research

This antibody is widely used in studies of MAPK signaling, cancer biology, and cellular regulation:

ApplicationOptimal DilutionKey Findings
ELISA1:5000–1:10,000 Quantifies phosphorylated RAF1 levels in cell lysates or serum
Immunofluorescence1:20–1:200 Visualizes subcellular localization (e.g., cytoplasm, mitochondria)
Western Blot1:500–1:2000 Identifies phosphorylation-dependent activation/inactivation of RAF1
Immunohistochemistry1:50–1:200 Maps RAF1 phosphorylation in tissue sections

Phosphorylation Context and Biological Significance

Phosphorylation at S43 is part of a complex regulatory network:

Phosphorylation SiteRegulatory ImpactCatalyst
S43Inactivation of RAF1 kinase activity MAPK1/ERK2
S338/S339Activation via PAK1/PAK5, enabling mitochondrial localization PAK1/PAK5
S621Stabilizes RAF1, preventing proteasomal degradation Growth factor signals

Dysregulation of S43 phosphorylation is linked to oncogenesis, as RAF1 hyperactivation drives uncontrolled cell proliferation .

Cancer Biology

  • Oncogenic Signaling: Phospho-RAF1 (S43) antibodies reveal RAF1’s role in MAPK-driven cancers, such as melanoma and colorectal cancer .

  • Therapeutic Targets: Detection of S43 phosphorylation aids in monitoring kinase inhibitor efficacy (e.g., MEK/ERK pathway inhibitors) .

Cellular Regulation

  • Apoptosis and Survival: S43 phosphorylation by ERK2 inhibits RAF1’s pro-survival functions, promoting apoptosis .

  • Mitochondrial Localization: Phosphorylation at S338/S339 (not S43) directs RAF1 to mitochondria, where it binds BCL2 to prevent cell death .

Product Specs

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

The Phospho-RAF1 (S43) Recombinant Monoclonal Antibody is meticulously generated through a well-defined process. The antibody genes are isolated from rabbits previously immunized with a synthesized peptide derived from the human RAF1 protein phosphorylated at S43. These genes are subsequently introduced into expression vectors and transfected into host suspension cells. Positive cells are then cultured to facilitate robust expression and secretion of the antibody. Following this, the Phospho-RAF1 (S43) Recombinant Monoclonal Antibody undergoes a comprehensive purification process using affinity chromatography, effectively separating the antibody from the surrounding cell culture supernatant. Finally, the antibody's functionality is thoroughly validated through ELISA and IF assays, confirming its ability to specifically interact with the human RAF1 protein phosphorylated at S43.

Phosphorylation of RAF1 at S43 plays a critical role in regulating the MAPK signaling pathway, impacting crucial cellular processes such as growth, survival, and differentiation. Dysregulation of this phosphorylation event can have significant consequences in cancer and other diseases characterized by aberrant signaling.

Form
Liquid
Lead Time
Typically, we can dispatch products within 1-3 working days after receiving your order. Delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
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, a serine/threonine-protein kinase, acts as a key regulatory link between membrane-associated Ras GTPases and the MAPK/ERK cascade. This crucial regulatory role acts as a switch determining cell fate decisions, including proliferation, differentiation, apoptosis, survival, and oncogenic transformation. RAF1 activation initiates a mitogen-activated protein kinase (MAPK) cascade, involving the sequential phosphorylation of the dual-specific MAPK kinases (MAP2K1/MEK1 and MAP2K2/MEK2) and the extracellular signal-regulated kinases (MAPK3/ERK1 and MAPK1/ERK2).

The phosphorylated form of RAF1 (on residues Ser-338 and Ser-339, by PAK1) phosphorylates BAD/Bcl2-antagonist of cell death at 'Ser-75'. It also phosphorylates adenylyl cyclases: ADCY2, ADCY5 and ADCY6, resulting in their activation. Additionally, RAF1 phosphorylates PPP1R12A, leading to inhibition of the phosphatase activity. Further, it phosphorylates TNNT2/cardiac muscle troponin T. RAF1 can promote NF-kB activation and inhibit signal transducers involved in motility (ROCK2), apoptosis (MAP3K5/ASK1 and STK3/MST2), proliferation and angiogenesis (RB1). RAF1 can also protect cells from apoptosis by translocating to the mitochondria where it binds BCL2 and displaces BAD/Bcl2-antagonist of cell death.

RAF1 regulates Rho signaling and migration and is essential for normal wound healing. It also plays a role in the oncogenic transformation of epithelial cells by repressing the TJ protein, occludin (OCLN), through the up-regulation of a transcriptional repressor SNAI2/SLUG. This leads to the down-regulation of OCLN. Notably, RAF1 restricts caspase activation in response to specific stimuli, including Fas stimulation, pathogen-mediated macrophage apoptosis, and erythroid differentiation.

Gene References Into Functions
  1. The functional assessment supported the pathogenicity of the RAF1 and RIT1 VUSs, while the significance of two variants of unknown significance in A2ML1 remained unclear. PMID: 29402968
  2. Our report presents the second familial case of Noonan syndrome due to a germline p.S427G substitution in RAF1 with no occurrence of a malignant tumor. This may suggest that carrying a germline mutation in the RAF1 oncogene is not associated with an increased risk of tumor development. It's important to note that RAF1 mutations have been observed as a somatic event in many types of cancer. PMID: 30204961
  3. Data indicate that Raf-1 proto-oncogene, serine-threonine kinase (RAF1) is a negative regulator of hepatocarcinogenesis. PMID: 28000790
  4. We report a patient with an inherited RAF1-associated Noonan syndrome, presenting with an antenatally diagnosed abnormality of skull shape, bilateral subdural haematomas of unknown cause, delayed myelination and polymicrogyria. PMID: 27753652
  5. Raf1 may serve as a novel prognostic factor and potential target for improving the long-term outcome of nonsmall cell lung cancer (NSCLC). PMID: 29484414
  6. Results provide evidence that RAF1 binding to SPRY4 is regulated by miR-1908 in glioma tumors. PMID: 29048686
  7. High RAF1 expression is associated with malignant melanoma. PMID: 28677804
  8. Two premature neonates with progressive biventricular hypertrophy found to have RAF1 variants in the CR2 domain are reported. PMID: 28777121
  9. Data indicate connector enhancer of kinase suppressor of Ras 1 protein (CNK1) as a molecular platform that controls c-raf protein (RAF) and c-akt protein (AKT) signaling and determines cell fate decisions in a cell type- and cell stage-dependent manner. PMID: 27901111
  10. CRAF is a bona fide alternative oncogene for BRAF/NRAS/GNAQ/GNA11 wild type melanomas PMID: 27273450
  11. Authors evaluated the expression of known targets of miR-125a and found that sirtuin-7, matrix metalloproteinase-11, and c-Raf were up-regulated in tumor tissue by 2.2-, 3-, and 1.7-fold, respectively. Overall, these data support a tumor suppressor role for miR-125a. PMID: 28445974
  12. Overexpression of ciRS-7 in HCT116 and HT29 cells led to the blocking of miR-7 and resulted in a more aggressive oncogenic phenotype, and ciRS-7 overexpression permitted the inhibition of miR-7 and subsequent activation of EGFR and RAF1 oncogenes PMID: 28174233
  13. miR-497 could serve as a tumor suppressor and a potential early diagnostic marker of gastric cancer by targeting Raf-1 proto-oncogene. PMID: 28586056
  14. RAF1 may have a role in survival in hepatocellular carcinoma, and indicate whether sorafenib should be used as a postoperative adjuvant PMID: 26981887
  15. Mutational activation of Kit-, Ras/Raf/Erk- and Akt- pathways indicate the biological importance of these pathways and their components as potential targets for therapy. PMID: 27391150
  16. Results indicate that des-gamma-carboxy prothrombin (DCP) antagonizes the inhibitory effects of Sorafenib on hepatocellular carcinoma (HCC) through activation of the Raf/MEK/ERK and PI3K/Akt/mTOR signaling 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 pathway are responsible for TNF-alpha-mediated endothelial cytotoxicity via regulation of the vimentin cytoskeleton. PMID: 28743511
  19. Although Raf-1 gene is not mutated, an abnormality of Raf-1 kinase feedback regulation enhances its antiapoptotic function, and Raf-1 can still be a pharmaceutical target to increase chemotherapy or radiotherapy sensitivity in these cancer cells. PMID: 27841865
  20. RAF1 plays a critical role in maintaining the transformed phenotype of CRC cells, including those with mutated KRAS. PMID: 27670374
  21. This finding suggests that stringent assemblage of Hsp90 keeps CRAF kinase equipped for participating in the MAPK pathway. Thus, the role of Hsp90 in CRAF maturation and activation acts as a limiting factor to maintain the function of a strong client like CRAF kinase. PMID: 27703006
  22. Oncogenic NFIA:RAF1 fusion activation of the MAPK pathway is associated with pilocytic astrocytoma. PMID: 27810072
  23. IGF2BP2 as a post-transcriptional regulatory mRNA-binding factor, interfering with Raf-1 degradation by miR-195, that contributes to Colorectal carcinogenesis. PMID: 27153315
  24. Data show that when microRNA miR-125b was over-expressed in THP-1 macrophages, the expression of Raf1 proto-oncogene serine/threonine protein kinase (RAF1) was reduced to promote the apoptosis of macrophages. PMID: 27363278
  25. Data show that Griffipavixanthone (GPX), a dimeric xanthone isolated from Garcinia esculenta, is a B-RAF and C-RAF inhibitor against esophageal cancer cells. PMID: 26646323
  26. Up-regulation of Raf-1 is associated with triple-negative breast cancer. PMID: 26513016
  27. This study provides the molecular basis for C-Raf C-terminal-derived phosphopeptide interaction with 14-3-3zeta protein and gives structural insights responsible for phosphorylation-mediated protein binding. PMID: 26295714
  28. a model that CD166 regulates MCAM through a signaling flow from activation of PI3K/AKT and c-Raf/MEK/ERK signaling to the inhibition of potential MCAM ubiquitin E3 ligases, betaTrCP and Smurf1. PMID: 26004137
  29. Suggest an interrelated kinase module involving c-Raf/PI3K/Lyn and perhaps Fgr functions in a nontraditional way during retinoic acid-induced maturation or during rescue of RA induction therapy using inhibitor co-treatment in RA-resistant leukemia cells. PMID: 25817574
  30. Abnormal activation of the Ras/MAPK pathway may play a significant role in the development of pulmonary vascular disease in the subset of patients with Noonan syndrome and a specific RAF1 mutation. PMID: 25706034
  31. Raf-1 may be an important biomarker in predicting the prognosis of chordoma patients. PMID: 25755752
  32. In the presence of Raf1, the RasQ61L mutant has a rigid switch II relative to the wild-type and increased flexibility at the interface with switch I, which propagates across Raf-Ras binding domain. PMID: 25684575
  33. Besides mediating the anticancer effect, pDAPK(S308) may serve as a predictive biomarker for Raf inhibitors combination therapy, suggesting an ideal preclinical model that is worthy of clinical translation. PMID: 26100670
  34. DJ-1 directly binds to the kinase domain of c-Raf to stimulate its self-phosphorylation, followed by phosphorylation of MEK and ERK1/2 in EGF-treated cells. PMID: 26048984
  35. Truncated RAF1 and BRAF proteins, recently described as products of genomic rearrangements in gastric cancer and other malignancies, have the ability to render neoplastic cells resistant to RTK-targeted therapy PMID: 25473895
  36. Our study demonstrated that miR-455-RAF1 may represent a new potential therapeutic target for colorectal carcinoma treatment. PMID: 25355599
  37. This approach identified 18 kinase and kinase-related genes whose overexpression can substitute for EGFR in EGFR-dependent PC9 cells, and these genes include seven of nine Src family kinase genes, FGFR1, FGFR2, ITK, NTRK1, NTRK2, MOS, MST1R, and RAF1. PMID: 25512530
  38. Aberrant expression of A-, B-, and C-RAF, and COT is frequent in PTC; increased expression of COT is correlated with recurrence of PTC. PMID: 25674762
  39. Authors demonstrate that the N-terminus of human Raf1 kinase (hRaf11-147aa) binds with human RKIP (hRKIP) at its ligand-binding pocket, loop "127-149", and the C-terminal helix by nuclear magnetic resonance experiments. PMID: 24863296
  40. Including several anti-apoptotic Bcl-2 family members and c-Raf. PMID: 24969872
  41. These data suggest that miR-7-5p functions as a tumor suppressor gene to regulate glioblastoma microvascular endothelial cell proliferation potentially by targeting the RAF1 oncogene PMID: 25027403
  42. A novel mechanism for response was discovered whereby high expression level of CAV-1 at the plasma membrane disrupts the BRaf/CRaf heterodimer and thus inhibits the activation of MAPK pathway during dasatinib treatment. PMID: 24486585
  43. Results show that ubiquitination and levels of RAF-1 is controlled by both Shoc2 and HUWE1. PMID: 25022756
  44. Raf-1/JNK /p53/p21 pathway may be involved in apoptosis, and NFkappaB1 may play a possible role in inhibiting apoptosis. PMID: 22282237
  45. The higher expression of RAF1 mRNA and the activation of AKT/ERK proteins in vinorelbine-resistant non-small cell lung cancer cell lines may confer resistance to vinorelbine PMID: 24427333
  46. Analysis of RAF1 mutations in cohorts of South Indian, North Indian and Japanese patients with childhood-onset dilated cardiomyopathy PMID: 24777450
  47. Expression of miR-195 or knockdown of Raf-1 can similarly reduce tumor cell survival. PMID: 23760062
  48. We hypothesize a potential direct or indirect role for SRC, RAF1, PTK2B genes in neurotransmission and in central nervous system signaling processes. PMID: 24108181
  49. We identified multiple C-RAF mutations that produced biochemical and pharmacologic resistance in melanoma cell lines PMID: 23737487
  50. ARAF seems to stabilize BRAF:CRAF complexes in cells treated with RAF inhibitors and thereby regulate cell signaling in a subtle manner to ensure signaling efficiency PMID: 22926515

Show More

Hide All

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 its significance in cellular signaling?

RAF1 (also known as c-RAF) is a serine/threonine protein kinase that functions as a MAP kinase kinase kinase (MAP3K) in the Ras-RAF-MEK-ERK signaling cascade. It operates downstream of Ras family membrane-associated GTPases, to which it binds directly. Once activated, RAF1 phosphorylates and activates the dual specificity protein kinases MEK1 and MEK2, which subsequently phosphorylate and activate ERK1 and ERK2. These activated ERKs are pleiotropic effectors that play crucial roles in controlling gene expression involved in cell division, apoptosis, differentiation, and migration . RAF1's central position in this pathway makes it a critical protein for studying normal cell physiology and pathological conditions, particularly in cancer research where aberrant pathway activation is common.

What is the significance of the S43 phosphorylation site on RAF1?

S43 represents one of several key phosphorylation sites on the RAF1 protein that regulate its activity. It is considered to be one of the basal in vivo RAF1 phosphorylation sites, alongside S259, S621, and another yet unidentified site . S43 is primarily characterized as an inhibitory phosphorylation site that is targeted by Protein Kinase A (PKA), although its functional significance remains somewhat controversial in the scientific literature . In the context of the RAF1 regulatory mechanism, S43 phosphorylation is thought to be part of a complex network of phosphorylation events that collectively fine-tune RAF1 activity in response to various cellular stimuli and signaling contexts.

How are recombinant monoclonal antibodies against phospho-RAF1 (S43) generated?

The generation of phospho-RAF1 (S43) recombinant monoclonal antibodies follows a sophisticated process that begins with the isolation of antibody-coding genes from rabbits immunized with a synthetic phospho-peptide derived from human RAF1 protein phosphorylated at S43 . These genes are then cloned into specialized expression vectors and transfected into host suspension cells. After successful transfection, the cells are cultured to facilitate antibody expression and secretion. The antibodies undergo purification through affinity chromatography to separate them from the cell culture supernatant. Finally, their functionality is rigorously evaluated through multiple analytical techniques including ELISA and immunofluorescence (IF) to confirm their specific interaction with human RAF1 protein phosphorylated at S43 .

What are the common applications for phospho-RAF1 (S43) antibodies in research?

Phospho-RAF1 (S43) antibodies are valuable tools in several research applications focused on MAPK pathway signaling and regulation. The most common applications include:

  • Western Blotting (WB): Used at dilutions of 1:500-1:5000 to detect phosphorylated RAF1 in cell or tissue lysates .

  • Immunofluorescence (IF): Applied at dilutions of 1:20-1:200 to visualize the subcellular localization of phosphorylated RAF1 in fixed cells or tissues .

  • Immunohistochemistry (IHC): Employed at dilutions of 1:50-1:200 to detect phosphorylated RAF1 in tissue sections .

  • Enzyme-Linked Immunosorbent Assay (ELISA): Used to quantify phosphorylated RAF1 levels in biological samples .

These techniques allow researchers to investigate how various stimuli, inhibitors, or genetic modifications affect RAF1 phosphorylation status and consequently, MAPK pathway activity in different experimental systems.

How does S43 phosphorylation interact with other RAF1 phosphorylation sites to regulate kinase activity?

RAF1 regulation involves a complex interplay between multiple phosphorylation sites that can either activate or inhibit its kinase activity. S43 phosphorylation does not function in isolation but operates within a sophisticated regulatory network. When S43 is phosphorylated by PKA, it may interfere with RAF1's ability to interact with Ras, thereby inhibiting activation . This site works in concert with other regulatory phosphorylation sites:

  • S259: When phosphorylated, provides a binding site for 14-3-3 proteins and stabilizes RAF1 in an inactive conformation

  • S621: When phosphorylated, provides a second binding site for 14-3-3 proteins that is critical for RAF1 kinase activity

  • S338/Y341: Phosphorylation at these sites is crucial for RAF1 activation by oncogenic Ras and growth factors

Recent research suggests that different pools of RAF1 may exist within cells with distinct phosphorylation patterns, and the interplay between these sites determines whether RAF1 adopts an active or inactive conformation. Understanding how S43 phosphorylation influences or is influenced by these other phosphorylation events represents an important area for advanced investigation, particularly in contexts where aberrant RAF1 activation contributes to disease states .

What methodological considerations are critical when using phospho-RAF1 (S43) antibodies in multiplexed phosphoproteomic analyses?

When incorporating phospho-RAF1 (S43) antibodies into multiplexed phosphoproteomic analyses, researchers must address several critical methodological considerations:

  • Antibody Specificity Validation: Rigorous validation is essential to ensure the antibody detects only RAF1 phosphorylated at S43. Cross-reactivity with other phosphorylation sites on RAF1 or with phosphorylated residues on related proteins can confound results .

  • Phosphatase Inhibitor Optimization: Since phosphorylation is labile, optimal preservation of phosphorylation status requires careful selection and concentration of phosphatase inhibitors in lysis buffers.

  • Multiplexed Antibody Compatibility: When combining with other antibodies, potential interference must be addressed through:

    • Selection of antibodies raised in different host species

    • Use of isotype-specific secondary antibodies

    • Sequential detection strategies if using the same host species

  • Signal Normalization: A robust strategy includes normalization to total RAF1 levels using parallel samples or sequential probing with total RAF1 antibodies after stripping.

  • Quantification Methods: Comparative analysis requires standardized quantification, preferably using:

Quantification MethodAdvantagesLimitations
Fluorescence-based detectionWide dynamic range, multiplexing capabilityPotential channel bleed-through
ChemiluminescenceHigh sensitivityMore limited dynamic range
Mass spectrometry validationAbsolute quantificationRequires specialized equipment

Implementation of these methodological considerations ensures reliable detection and quantification of S43 phosphorylation in complex biological samples and experimental systems .

How can researchers distinguish between PKA-mediated and other kinase-mediated phosphorylation of RAF1 at S43?

Distinguishing the specific kinase responsible for S43 phosphorylation requires a multifaceted experimental approach:

  • Pharmacological Inhibition Studies: Selective inhibition of PKA using compounds such as H-89 or PKI, followed by assessment of S43 phosphorylation levels. A significant reduction would suggest PKA involvement, while persistence might indicate alternative kinases .

  • Genetic Manipulation Approaches:

    • siRNA/shRNA knockdown of PKA catalytic subunits

    • CRISPR-Cas9 mediated knockout of PKA genes

    • Expression of dominant-negative PKA mutants

    These interventions should be followed by quantitative assessment of S43 phosphorylation.

  • In Vitro Kinase Assays: Purified candidate kinases (PKA, PKC, etc.) can be tested for their ability to phosphorylate RAF1 peptides or recombinant proteins containing the S43 site, with phosphorylation detected by either:

    • Phospho-specific antibodies

    • Radioactive ATP incorporation

    • Mass spectrometry

  • Consensus Sequence Analysis: Examination of the amino acid sequence surrounding S43 for conformity to known kinase consensus motifs:

KinaseConsensus MotifMatch with S43 Region
PKAR-R/K-X-S/TStrong match
PKCS/T-X-R/KWeak/no match
AMPKΦ-X-B-X-X-S/T-X-X-X-ΦPartial match

Where Φ = hydrophobic residue, B = basic residue, X = any amino acid

  • Temporal Activation Patterns: Time-course experiments examining S43 phosphorylation following selective activation of different kinases can help discriminate between rapid direct effects and delayed indirect phosphorylation events .

Through this comprehensive approach, researchers can definitively attribute S43 phosphorylation to specific kinases in various physiological and pathological contexts.

What experimental controls are essential when validating specificity of phospho-RAF1 (S43) antibodies?

Validating the specificity of phospho-RAF1 (S43) antibodies requires a systematic approach with several critical controls:

  • Phosphatase Treatment Control: Treatment of one sample set with lambda phosphatase should eliminate the phospho-specific signal while preserving total RAF1 detection. This confirms the antibody's phospho-specificity .

  • Peptide Competition Assay: Pre-incubating the antibody with:

    • Phosphorylated S43 peptide (should block specific signal)

    • Non-phosphorylated S43 peptide (should not affect specific signal)

    • Phosphorylated peptides from other RAF1 sites (should not affect specific signal)

  • Genetic Controls:

    • S43A mutation (phospho-null): Should show no signal with the phospho-specific antibody

    • S43D/E mutation (phospho-mimetic): May show reduced or no signal depending on epitope recognition

  • Stimulus-Response Validation:

    • Treatment with PKA activators (e.g., forskolin, cAMP analogs) should increase signal

    • Treatment with PKA inhibitors should decrease signal

    • Stimuli known to modulate MAPK pathway (e.g., growth factors) may alter signal in a predictable manner

  • Cross-Reactivity Assessment: Testing the antibody against:

    • Recombinant phosphorylated and non-phosphorylated RAF1 protein

    • Lysates from cells expressing RAF1 family members (A-RAF, B-RAF) to assess isoform specificity

    • Knockout or knockdown cells lacking RAF1 expression

These controls, when properly implemented and documented, provide comprehensive validation of antibody specificity and ensure reliable experimental outcomes with phospho-RAF1 (S43) antibodies .

What are the optimal sample preparation protocols for preserving RAF1 S43 phosphorylation status?

Preserving the phosphorylation status of RAF1 at S43 during sample preparation requires careful attention to multiple factors:

  • Cell/Tissue Harvesting:

    • Rapid processing is essential as phosphorylation states can change within seconds

    • For adherent cells, direct lysis in the culture dish is preferable to trypsinization

    • For tissues, snap-freezing in liquid nitrogen immediately after collection is critical

  • Lysis Buffer Composition:

    • Robust phosphatase inhibitor cocktail containing sodium fluoride (50 mM), sodium orthovanadate (1 mM), β-glycerophosphate (10 mM), and sodium pyrophosphate (5 mM)

    • Protease inhibitors to prevent degradation of RAF1 protein

    • Non-denaturing detergents (NP-40 or Triton X-100, 0.5-1%) for applications requiring native protein

    • SDS-containing buffers (1-2%) for applications where denaturation is acceptable

  • Physical Parameters:

    • Maintain samples at 4°C throughout processing

    • Avoid repeated freeze-thaw cycles

    • Process samples immediately or store at -80°C with phosphatase inhibitors

  • Protein Concentration Determination:

    • Use methods compatible with phosphatase inhibitors (Bradford or BCA)

    • Dilute samples consistently to ensure comparable protein loading

  • Sample Storage:

    • Add reducing agents (DTT or β-mercaptoethanol) immediately before use

    • For long-term storage, aliquot samples to avoid repeated freeze-thaw cycles

    • Store at -80°C with phosphatase inhibitors

For specialized applications such as mass spectrometry analysis of phosphorylation sites, additional considerations include phosphopeptide enrichment using titanium dioxide (TiO₂) or immobilized metal affinity chromatography (IMAC) to enhance detection sensitivity of S43 phosphorylation .

How can quantitative analysis of RAF1 S43 phosphorylation be standardized across different experimental systems?

Standardizing quantitative analysis of RAF1 S43 phosphorylation across different experimental systems requires a systematic approach to ensure consistency and reproducibility:

  • Reference Standards Implementation:

    • Include recombinant phosphorylated RAF1 protein standards at known concentrations

    • Develop a standard curve with varying ratios of phosphorylated to total RAF1

    • Consider synthetic phosphopeptide standards for mass spectrometry applications

  • Normalization Strategy:

    • Always normalize phospho-S43 signal to total RAF1 protein levels

    • Include housekeeping proteins (GAPDH, actin, tubulin) as loading controls

    • Consider multiple normalization references to ensure robustness

  • Quantification Methodology:

MethodApplicationQuantification Approach
Western BlotProtein lysatesDensitometry with linear dynamic range validation
ELISASoluble samplesFour-parameter logistic regression standard curve
Flow CytometrySingle cellsMedian fluorescence intensity with isotype controls
ImmunofluorescenceFixed cells/tissuesIntegrated intensity per cell with background subtraction
Mass SpectrometryPeptide digestsHeavy isotope-labeled internal standards
  • Inter-laboratory Validation:

    • Exchange of positive control samples between laboratories

    • Implementation of standard operating procedures (SOPs)

    • Blind analysis of identical samples by different researchers

  • Data Reporting Requirements:

    • Always report phospho-S43:total RAF1 ratios rather than absolute values

    • Include positive controls (PKA activator treatment) and negative controls (PKA inhibitor treatment)

    • Document all normalization procedures in detail

    • Report biological and technical replicate numbers with appropriate statistics

By implementing these standardization approaches, researchers can achieve consistent and comparable quantification of RAF1 S43 phosphorylation across different experimental systems, instruments, and laboratories .

How should researchers interpret discrepancies between phospho-RAF1 (S43) levels and functional outcomes in the MAPK pathway?

Interpreting discrepancies between phospho-RAF1 (S43) levels and MAPK pathway activation requires careful consideration of several biological complexities:

  • Multi-site Phosphorylation Effects:

    • S43 phosphorylation represents just one of many regulatory modifications on RAF1

    • The net effect on RAF1 activity depends on the combination of phosphorylation at multiple sites including S259, S338, Y341, and S621

    • Analyze phosphorylation at these additional sites simultaneously to obtain a complete regulatory picture

  • Temporal Dynamics:

    • S43 phosphorylation may exhibit different kinetics than downstream MAPK pathway activation

    • Conduct comprehensive time-course experiments to correlate S43 phosphorylation with:

      • MEK phosphorylation

      • ERK phosphorylation

      • ERK-dependent transcriptional responses

  • Compartmentalization Considerations:

    • Investigate subcellular localization of phospho-S43 RAF1 using immunofluorescence or subcellular fractionation

    • Different pools of RAF1 (membrane-associated, cytoplasmic, nuclear) may have distinct signaling properties

    • Phospho-S43 RAF1 in specific compartments may correlate better with functional outcomes

  • Threshold Effects:

    • The relationship between phosphorylation and pathway activation may be non-linear

    • Determine if a threshold level of S43 phosphorylation is required to observe inhibitory effects

    • Titrate activators/inhibitors to establish dose-response relationships

  • Competing Regulatory Mechanisms:

    • Other proteins in the MAPK pathway (e.g., scaffold proteins, phosphatases) may compensate for S43 phosphorylation

    • Investigate additional regulatory mechanisms that might override S43 phosphorylation effects

    • Consider the role of RAF dimerization, which may be affected by S43 phosphorylation

What are the most common sources of false positive and false negative results when detecting phospho-RAF1 (S43), and how can they be mitigated?

Detection of phospho-RAF1 (S43) can be complicated by several factors that lead to false results. Understanding and mitigating these factors is crucial for accurate data interpretation:

Common Sources of False Positives and Mitigation Strategies:

  • Cross-reactivity with Other Phosphorylation Sites:

    • Cause: Antibodies may recognize similar phosphorylated motifs on RAF1 or related proteins

    • Mitigation: Validate antibody specificity using phospho-null mutants (S43A) and peptide competition assays

  • Phosphorylation During Sample Handling:

    • Cause: Stress during cell harvesting can activate kinases that phosphorylate S43

    • Mitigation: Rapid lysis with phosphatase and kinase inhibitors; process samples at 4°C

  • Non-specific Binding in Immunoassays:

    • Cause: High antibody concentrations or insufficient blocking

    • Mitigation: Optimize antibody dilutions; use proper blocking agents; include IgG controls

  • Artifactual Phosphorylation in Fixed Samples:

    • Cause: Some fixatives can create epitopes resembling phosphorylated residues

    • Mitigation: Compare multiple fixation methods; validate with alternative detection methods

Common Sources of False Negatives and Mitigation Strategies:

  • Rapid Dephosphorylation:

    • Cause: Phosphatases actively remove phosphate groups during sample preparation

    • Mitigation: Use comprehensive phosphatase inhibitor cocktails; maintain cold temperatures

  • Epitope Masking:

    • Cause: Protein interactions or conformational changes may hide the S43 phospho-epitope

    • Mitigation: Test multiple lysis conditions; consider denaturing conditions for Western blotting

  • Insufficient Sensitivity:

    • Cause: Low abundance of phosphorylated form relative to total RAF1

    • Mitigation: Enrich phosphoproteins using phospho-specific immunoprecipitation; use signal amplification methods; employ more sensitive detection systems

  • Timing of Analysis:

    • Cause: S43 phosphorylation may be transient

    • Mitigation: Perform detailed time-course experiments; synchronize cells if appropriate

Comprehensive Validation Approach:

TechniqueControls for False PositivesControls for False Negatives
Western BlotPhosphatase treatment; peptide competitionPositive control (PKA activator treatment)
ImmunofluorescenceSecondary antibody only; pre-immune serumPermeabilization optimization; signal amplification
ELISAStandard curve; non-phospho peptide controlsPhosphatase inhibitors; spike-in controls
Mass SpectrometryDecoy database searchesPhosphopeptide enrichment; internal standards

By implementing these mitigation strategies, researchers can significantly reduce false results and increase confidence in phospho-RAF1 (S43) detection across various experimental platforms .

How can researchers effectively use phospho-RAF1 (S43) antibodies to investigate crosstalk between MAPK and other signaling pathways?

Investigating signaling crosstalk using phospho-RAF1 (S43) antibodies requires sophisticated experimental designs that capture the integration of multiple pathways:

  • Dual Pathway Stimulation/Inhibition Experiments:

    • Approach: Systematically activate or inhibit candidate pathways while monitoring S43 phosphorylation

    • Design Example:

Treatment ConditionMAPK Pathway ActivatorInteracting Pathway ModulatorExpected Effect on S43 Phosphorylation
Control--Baseline
MAPK activationEGF/Serum-Pathway-specific response
Interacting pathway-PKA activator (forskolin)Increase if direct relationship
Dual activationEGF/SerumPKA activator (forskolin)Synergistic or antagonistic effect reveals crosstalk
Pathway inhibitionMEK inhibitorPKA inhibitorReveals dependency relationships
  • Temporal Resolution of Pathway Activation:

    • Approach: Detailed time-course experiments with multiple pathway readouts

    • Implementation: Harvest cells at closely spaced time points (0, 5, 15, 30, 60 min, etc.) after stimulation

    • Analysis: Compare phosphorylation kinetics of:

      • RAF1 S43 phosphorylation

      • RAF1 activation markers (S338 phosphorylation)

      • Downstream MAPK components (phospho-MEK, phospho-ERK)

      • Readouts from interacting pathways (phospho-CREB for PKA, phospho-AKT for PI3K)

  • Genetic Perturbation Combined with Biochemical Analysis:

    • Approach: Modify components of interacting pathways and assess impact on S43 phosphorylation

    • Implementation:

      • CRISPR knockout or knockdown of PKA catalytic subunits

      • Expression of constitutively active or dominant-negative pathway components

      • Rescue experiments with wild-type or phospho-mutant RAF1 (S43A)

    • Readout: Multi-parameter analysis including phospho-S43 RAF1, pathway activity markers, and functional outcomes

  • Spatial Organization Analysis:

    • Approach: Investigate subcellular compartmentalization of signaling components

    • Techniques:

      • Confocal microscopy with co-localization analysis of phospho-S43 RAF1 with components of other pathways

      • Proximity ligation assay to detect protein-protein interactions in situ

      • Subcellular fractionation followed by Western blotting for pathway components

  • Systems-Level Analysis:

    • Approach: Global phosphoproteomics combined with targeted validation

    • Implementation:

      • SILAC or TMT-based phosphoproteomics under various pathway perturbation conditions

      • Bioinformatic analysis to identify coordinated phosphorylation events

      • Validation of key nodes with phospho-specific antibodies including phospho-S43 RAF1

This multi-faceted approach allows researchers to dissect how S43 phosphorylation serves as an integration point for multiple signaling inputs and how this ultimately affects RAF1 function in complex cellular contexts .

What are the future directions for research using phospho-RAF1 (S43) antibodies in understanding signaling dynamics and disease mechanisms?

The continuing evolution of research employing phospho-RAF1 (S43) antibodies presents several promising directions for advancing our understanding of signaling dynamics and disease mechanisms:

  • Single-Cell Analysis of Phosphorylation Heterogeneity:
    Future studies will likely leverage phospho-RAF1 (S43) antibodies in single-cell technologies to understand cell-to-cell variability in RAF1 regulation. This approach will reveal how phosphorylation heterogeneity contributes to diverse cellular responses within populations and how this heterogeneity may influence disease progression, particularly in cancer where MAPK pathway dysregulation is common .

  • Spatiotemporal Dynamics of RAF1 Phosphorylation:
    Development of new biosensors and live-cell imaging techniques incorporating phospho-RAF1 (S43) detection will enable real-time visualization of phosphorylation dynamics. This will provide unprecedented insights into how rapidly S43 phosphorylation occurs in response to various stimuli and how it correlates with subcellular localization and RAF1 activation state .

  • Integration with Multi-omics Approaches:
    Combining phospho-RAF1 (S43) antibody-based detection with proteomics, transcriptomics, and metabolomics will create comprehensive models of how S43 phosphorylation affects global cellular states. This systems biology approach will help decipher the full spectrum of downstream effects resulting from alterations in S43 phosphorylation status .

  • Therapeutic Target Validation:
    As RAF inhibitors continue to be developed for cancer treatment, understanding the role of S43 phosphorylation in drug response and resistance will be crucial. Phospho-RAF1 (S43) antibodies will serve as important tools for monitoring pathway adaptations during treatment and for identifying combination therapy strategies that might overcome resistance mechanisms .

  • Development of Phosphorylation-State Specific Modulators:
    Knowledge gained from studies using phospho-RAF1 (S43) antibodies may guide the development of novel therapeutics that specifically target or exploit the S43 phosphorylation state. Such approaches could offer more precise manipulation of RAF1 activity compared to current inhibitors that target the kinase domain .

As methods for detecting and quantifying phosphorylation events become more sophisticated, our understanding of how S43 phosphorylation contributes to normal physiology and disease states will continue to deepen, potentially opening new avenues for therapeutic intervention in MAPK pathway-driven diseases .

How can phospho-RAF1 (S43) antibodies contribute to the development of personalized medicine approaches for RAF-pathway driven diseases?

Phospho-RAF1 (S43) antibodies offer significant potential for advancing personalized medicine in diseases driven by aberrant RAF pathway activation:

  • Biomarker Development for Treatment Selection:
    Phospho-RAF1 (S43) status could serve as a predictive biomarker for response to various targeted therapies. By analyzing S43 phosphorylation in patient samples using validated immunohistochemical protocols, clinicians might better stratify patients for:

    • RAF inhibitor therapy

    • MEK inhibitor therapy

    • Combination approaches

    • Alternative pathway inhibitors when S43 phosphorylation indicates specific crosstalk mechanisms

  • Monitoring Treatment Response and Resistance Development:
    Serial assessment of phospho-RAF1 (S43) levels during treatment could provide early indicators of:

    • Initial treatment efficacy before clinical response is apparent

    • Development of adaptive resistance

    • Pathway reactivation requiring treatment modification

    This monitoring could enable more dynamic treatment adjustments based on molecular changes rather than waiting for clinical progression .

  • Identification of Patient-Specific Pathway Dysregulation:
    Analysis of phospho-RAF1 (S43) in conjunction with other pathway components could reveal patient-specific patterns of dysregulation:

Phosphorylation PatternPotential Therapeutic Implication
High p-S43/Low p-S338PKA-mediated inhibition dominant; consider PKA modulation
Low p-S43/High p-S338Constitutive activation; RAF or MEK inhibitors indicated
High p-S43/High p-S338Pathway resistance to normal regulation; consider multiple targeting
Variable p-S43 in tumor regionsHeterogeneous disease; combination therapy may prevent resistance
  • Development of Functional Diagnostic Assays:
    Ex vivo testing of patient-derived samples could evaluate dynamic changes in S43 phosphorylation in response to candidate therapeutics. This functional diagnostics approach would provide personalized prediction of drug efficacy before initiating treatment .

  • Integration with Genomic Medicine:
    Correlating S43 phosphorylation status with specific genetic alterations could enhance the interpretative value of genomic testing. This integration would create more comprehensive predictive models that consider both genetic drivers and their consequent phosphorylation states .

  • Therapeutic Resistance Mechanisms:
    Understanding how alterations in S43 phosphorylation contribute to therapy resistance could inform secondary treatment strategies. Phospho-RAF1 (S43) antibodies could help identify specific resistance mechanisms related to:

    • Feedback activation of upstream signals

    • Altered RAF dimerization

    • Pathway crosstalk compensation

    • Phosphatase upregulation

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.