Phospho-MAP2K1 (T292) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

CUSABIO designed the vector clones for the expression of a recombinant MAP2K1 antibody in mammalian cells. The vector clones were obtained by inserting the MAP2K1 antibody heavy and light chains into the plasma vectors. The recombinant MAP2K1 antibody was purified from the culture medium through affinity-chromatography. It can be used to detect MAP2K1 protein from Human in the ELISA, WB.

The phospho-MAP2K1 (T292) antibody can detect the MAP2K1 only when phosphorylated at T292. MAP2K1, also called MEK1, functions immediately upstream of MAPK in the MAPK signaling pathway. Phosphorylation of T292 on MAP2K1 by activated ERK is required for the formation of ternary complex PTEN/MAGI1/MAP2K1. MAP2K1 mutations have been found in several human malignancies, particularly melanoma, hairy cell leukemia, and lung adenocarcinoma.

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 MAP2K1 antibody in mammalian cells. These clones were generated by inserting the MAP2K1 antibody heavy and light chains into suitable plasma vectors. The recombinant MAP2K1 antibody was subsequently purified from the culture medium using affinity-chromatography. This antibody is designed for the detection of MAP2K1 protein from Human in ELISA and Western blotting applications.

The phospho-MAP2K1 (T292) antibody exhibits specificity for MAP2K1 only when phosphorylated at T292. MAP2K1, also known as MEK1, plays a pivotal role as an upstream regulator of MAPK within the MAPK signaling pathway. Phosphorylation of T292 on MAP2K1 by activated ERK is essential for the formation of a ternary complex composed of PTEN, MAGI1, and MAP2K1. Notably, mutations in MAP2K1 have been implicated in various human malignancies, particularly melanoma, hairy cell leukemia, and lung adenocarcinoma.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 working days after receiving your order. The delivery time may vary depending on the chosen shipping method or destination. We recommend contacting your local distributor for specific delivery timelines.
Synonyms
Dual specificity mitogen activated protein kinase kinase 1 antibody; Dual specificity mitogen-activated protein kinase kinase 1 antibody; ERK activator kinase 1 antibody; MAP kinase kinase 1 antibody; MAP kinase/Erk kinase 1 antibody; MAP2K1 antibody; MAPK/ERK kinase 1 antibody; MAPKK 1 antibody; MAPKK1 antibody; MEK 1 antibody; Mek1 antibody; MEKK1 antibody; Mitogen activated protein kinase kinase 1 antibody; MKK 1 antibody; MKK1 antibody; MP2K1_HUMAN antibody; PRKMK1 antibody; Protein kinase mitogen activated kinase 1 (MAP kinase kinase 1) antibody; Protein kinase mitogen activated, kinase 1 antibody; protein kinase mitogen-activated kinase 1 antibody
Target Names
Uniprot No.

Target Background

Function

MAP2K1 (also known as MEK1) is a dual specificity protein kinase that serves as a critical component of the MAP kinase signal transduction pathway. The activation of this pathway is initiated by the binding of extracellular ligands, such as growth factors, cytokines, and hormones, to their corresponding cell-surface receptors. This binding event triggers the activation of RAS, which in turn activates RAF1. RAF1 then further activates the dual-specificity protein kinases MAP2K1/MEK1 and MAP2K2/MEK2. Both MAP2K1/MEK1 and MAP2K2/MEK2 are specifically involved in the MAPK/ERK cascade. They catalyze the phosphorylation of a threonine and a tyrosine residue within a Thr-Glu-Tyr sequence located in the extracellular signal-regulated kinases MAPK3/ERK1 and MAPK1/ERK2. This phosphorylation event leads to the activation of these kinases, allowing for further signal transduction within the MAPK/ERK cascade.

MAP2K1/MEK1 also activates BRAF in a KSR1 or KSR2-dependent manner. Binding to KSR1 or KSR2 releases the inhibitory intramolecular interaction between the KSR1 or KSR2 protein kinase and N-terminal domains, which promotes KSR1 or KSR2-BRAF dimerization and subsequent activation of BRAF. Depending on the cellular context, the MAPK/ERK cascade orchestrates a wide range of biological functions, including cell growth, adhesion, survival, and differentiation. These functions are primarily mediated through the regulation of transcription, metabolism, and cytoskeletal rearrangements. One target of the MAPK/ERK cascade is peroxisome proliferator-activated receptor gamma (PPARG), a nuclear receptor that promotes differentiation and apoptosis. MAP2K1/MEK1 has been shown to export PPARG from the nucleus. The MAPK/ERK cascade also participates in the regulation of endosomal dynamics, including lysosome processing and endosome cycling through the perinuclear recycling compartment (PNRC), as well as the fragmentation of the Golgi apparatus during mitosis.

Gene References Into Functions
  1. Upregulation of miR101 inhibited DLBCL cell proliferation and facilitated apoptosis by inhibiting the expression of MEK1. PMID: 30365139
  2. Somatic activating mutations in MAP2K1 cause melorheostosis. PMID: 29643386
  3. A direct interaction of both MEK1 and MEK2 with AKT has been identified. This interaction between MEK and AKT affects cell migration and adhesion, but not proliferation. The specific mechanism of action of the MEK-AKT complex involves phosphorylation of the migration-related transcription factor FoxO1. PMID: 28225038
  4. Activating mutations enhance the rate of MEK1 phosphorylation by Raf. These mutations also affect the thermal stability of MEK1 and its activity towards ERK2. PMID: 29018093
  5. Two atypical hairy cell leukemia (HCLc)- and hairy cell leukemia variant (HCLv)-like cases with clinically detected mitogen-activated protein kinase kinase 1 (MAP2K1) mutations have been reported. PMID: 27241017
  6. Reactive oxygen species-mediated EGFR/MEK/ERK/HIF-1A loop regulates glucose metabolism in pancreatic cancer. PMID: 29702094
  7. High MEK1 expression is associated with urinary bladder cancer metastasis. PMID: 28534984
  8. Mutually exclusive KRAS and MAP2K1 mutations were detected in one-third of cases of Rosai-Dorfman disease. PMID: 28664935
  9. The rs1549854 and rs1432441 polymorphisms of the MAP2K1 gene may be associated with major depressive disorder, especially in females. PMID: 28688265
  10. MEK1 does not act as a general tumor suppressor in leukemogenesis. Its effects are strongly dependent on the genetic context (RAS versus MYC-driven leukemia) and on the cell type involved. PMID: 27741509
  11. A synthetic lethal interaction of cetuximab in combination with MEK1/2 inhibition for the NRAS mutant subgroup of metastatic colorectal cancer has been reported. PMID: 27636997
  12. High MEK1 expression is associated with liver cancer. PMID: 26967560
  13. BRAF/MAP2K1-mut LCH cells exhibited a more immature state than BRAF/MAP2K1-wt LCH cells. The BRAFV600E and MAP2K1 mutations were significantly associated with pERK expression. PMID: 27597420
  14. There are no other biomarkers correlated with treatment responses following MEK1/2 inhibition. PMID: 27956260
  15. High MEK1 expression is associated with neuroblastoma. PMID: 28687621
  16. Mutations in MAP2K1, which are frequently associated with neurological complications and intellectual disability, can be associated with a milder clinical and neurocognitive profile more typical of individuals with Noonan syndrome. Variability of expression may arise from a complex interplay between RAS/MAPK pathway genotype, epigenetics, medical and obstetric factors, and environmental influences. PMID: 27862862
  17. High MEK1 expression is associated with infant acute lymphoblastic leukemia. PMID: 27588400
  18. Combined therapy using a HER2 inhibitor and a BRAF/MEK inhibitor presented a more significant redifferentiation effect on papillary thyroid cancer cells harboring BRAFV600E than BRAF/MEK inhibitor alone. PMID: 28423638
  19. MEK1 is constitutively and mainly phosphorylated at the Thr-292, Ser-298, Thr-386, and Thr-388 residues in vivo. Combinations of phosphorylations at these four residues produce at least six phosphorylated variants of MEK1. The phosphorylation statuses of Thr-292, Ser-298, Thr-386, and Thr-388 residues vary widely during activation and deactivation of the MAPK pathway. PMID: 27169363
  20. TNFRSF14 and MAP2K1 mutations are the most frequent genetic alterations found in pediatric-type follicular lymphoma (PTFL) and occur independently in most cases, suggesting that both mutations might play an important role in PTFL lymphomagenesis. PMID: 28533310
  21. There was no statistically significant association between BRAF or MAP2K1 mutation and anatomic site, unifocal versus multifocal presentation, or clinical outcome in Langerhans cell histiocytosis. PMID: 26980021
  22. High MEK1 expression is associated with inflammation. PMID: 28178421
  23. Lgr4 is a critical positive factor for skin tumorigenesis by mediating the activation of MEK1/ERK1/2 and Wnt/beta-catenin pathways. PMID: 27693558
  24. Somatic mutations in MAP2K1 are a common cause of extracranial arteriovenous malformation. PMID: 28190454
  25. MEK1 mutation is associated with central nervous system metastases of non-small cell lung cancer. PMID: 26860843
  26. The MAP2K1 mutation analysis of three hairy cell leukemia cases, one hairy cell leukemia-variant case, and three splenic marginal zone lymphoma cases revealed negative results. PMID: 25729732
  27. Data show that mitogen-activated protein kinase kinases MEK1/2 inhibitor pimasertib (MEKI) sensitized the cells to apoptosis through its ability to promote a G1 cell cycle arrest. PMID: 26625317
  28. Specific inhibition of BRAF oncogene, MEK or p38 signaling was associated with decreases in DIO3 expression in papillary thyroid cancer cells. PMID: 26825960
  29. Data show that Ba/F3 cells transformed with mutant HRAS protien indicated equal sensitivity towards Map kinase kinase (MEK) and mTOR serine-threonine kinase (mTOR) inhibition. PMID: 26544513
  30. Our data demonstrate that MEK inhibitors can inhibit breast cancer stem cells and may have clinical potential for the prevention of metastasis in certain cases in which tumors are MAPK dependent. PMID: 26384399
  31. Data show that src kinases (SRC) and mitogen-activated protein kinase kinase 1 (MEK) co-inhibition by saracatinib and PD0325901 respectively can be broadly effective in tumor growth control of a wide panel of non-small cell lung cancer (NSCLC) cell lines. PMID: 26358373
  32. At clinically relevant concentrations, cDDP binds to and inhibits MEK1/2, and both the binding and inhibitory activity are related to its interaction with Cu bound to MEK1/2. PMID: 26155939
  33. Studies indicate that concurrent inhibition of proto-oncogene protein B-raf (BRAF) and Map kinase kinase (MEK) improved the most effective therapeutic modality as compared to single BRAF or MEK inhibition for patients with metastatic melanoma (MM). PMID: 26143635
  34. Findings suggest that triple therapy directed against BRAF/MEK/ErbB3 may be able to provide durable control of BRAF mutated metastatic melanoma. PMID: 26208478
  35. MEK1 levels are upregulated at the transcriptional level whereas MEK2 levels are downregulated at the posttranslational level. PMID: 26163823
  36. NOTCH1, TP53, and MAP2K1 mutations in splenic diffuse red pulp small B-cell lymphoma are associated with progressive disease. PMID: 26426381
  37. MEK1/2 inhibitor trametinib showed similar PFS and a response rate as docetaxel in patients with previously treated KRAS-mutant-positive non-small cell lung carcinoma. PMID: 25722381
  38. Findings support the hypothesis that BDNF and MEK1 mRNA expression levels are more obviously decreased in patients with treatment-resistant depression. PMID: 24709918
  39. MEK1/2 inhibitor potentiated the anti-tumor effects of cisplatin in KRAS-dependent lung cancer cells and an animal model through inhibition of BIM degradation. PMID: 25541062
  40. Treatment of cells with sirtuin inhibitors, or siRNA knockdown of SIRT1 or SIRT2 proteins, increases MEK1 acetylation and subsequent phosphorylation of the extracellular signal-regulated kinase. PMID: 24681949
  41. MEK1 is associated with carboplatin resistance and is a prognostic biomarker in epithelial ovarian cancer. PMID: 25408231
  42. We documented three novel mutations in the BRAF gene in cardio-facio-cutaneous syndrome patients and correlated clinical findings with causative mutations in the BRAF or MEK1/MEK2 genes. PMID: 25463315
  43. MAP2K1 missense mutations were found in 2 of 11 patients with cadiofaciocutaneous syndrome: Pro124Gln and Asp67Asn. PMID: 25194980
  44. our data indicate that preexisting MEK1(P124) mutations are associated with a reduced response to BRAF inhibitor therapy and identify a subset of patients with BRAF-mutant melanoma likely to benefit from combination therapies. PMID: 25370473
  45. Langerhans cell histiocytosis cells can harbor additional genetic alterations in the RAS-RAF-MEK pathway which, in the case of MAP2K1, may be responsible for ERK activation in a wild type BRAF setting. PMID: 25899310
  46. Data show that licochalcone A (LicoA) suppresses solar UV-induced cyclooxygenase (COX-2) expression by acting as a potent inhibitor of enzymes PI3K, MEK1, and B-Raf. PMID: 25710724
  47. MEK1 Mutations are associated with Low-grade Serous Ovarian Cancer. PMID: 26324360
  48. Findings establish that the convergence of 2 distinct Ras effector pathways on mammalian target of rapamycin signaling maintains neurofibromatosis type 1 mouse optic glioma growth. PMID: 25534823
  49. MEK1/2 inhibitor trametinib showed similar PFS and a response rate as docetaxel in patients with previously treated KRAS-mutant-positive non-small cell lung carcinoma. PMID: 25722381
  50. NOTCH1, TP53, and MAP2K1 mutations in splenic diffuse red pulp small B-cell lymphoma are associated with progressive disease. PMID: 26426381
  51. MEK1 levels are upregulated at the transcriptional level whereas MEK2 levels are downregulated at the posttranslational level. PMID: 26163823
  52. Findings suggest that triple therapy directed against BRAF/MEK/ErbB3 may be able to provide durable control of BRAF mutated metastatic melanoma. PMID: 26208478
  53. Studies indicate that concurrent inhibition of proto-oncogene protein B-raf (BRAF) and Map kinase kinase (MEK) improved the most effective therapeutic modality as compared to single BRAF or MEK inhibition for patients with metastatic melanoma (MM). PMID: 26143635
  54. At clinically relevant concentrations, cDDP binds to and inhibits MEK1/2, and both the binding and inhibitory activity are related to its interaction with Cu bound to MEK1/2. PMID: 26155939
  55. Data show that src kinases (SRC) and mitogen-activated protein kinase kinase 1 (MEK) co-inhibition by saracatinib and PD0325901 respectively can be broadly effective in tumor growth control of a wide panel of non-small cell lung cancer (NSCLC) cell lines. PMID: 26358373
  56. Our data demonstrate that MEK inhibitors can inhibit breast cancer stem cells and may have clinical potential for the prevention of metastasis in certain cases in which tumors are MAPK dependent. PMID: 26384399
  57. Data show that Ba/F3 cells transformed with mutant HRAS protien indicated equal sensitivity towards Map kinase kinase (MEK) and mTOR serine-threonine kinase (mTOR) inhibition. PMID: 26544513
  58. Specific inhibition of BRAF oncogene, MEK or p38 signaling was associated with decreases in DIO3 expression in papillary thyroid cancer cells. PMID: 26825960
  59. Data show that mitogen-activated protein kinase kinases MEK1/2 inhibitor pimasertib (MEKI) sensitized the cells to apoptosis through its ability to promote a G1 cell cycle arrest. PMID: 26625317
  60. The MAP2K1 mutation analysis of three hairy cell leukemia cases, one hairy cell leukemia-variant case, and three splenic marginal zone lymphoma cases revealed negative results. PMID: 25729732
  61. MEK1 mutation is associated with central nervous system metastases of non-small cell lung cancer. PMID: 26860843
  62. Somatic mutations in MAP2K1 are a common cause of extracranial arteriovenous malformation. PMID: 28190454
  63. Lgr4 is a critical positive factor for skin tumorigenesis by mediating the activation of MEK1/ERK1/2 and Wnt/beta-catenin pathways. PMID: 27693558
  64. High MEK1 expression is associated with inflammation. PMID: 28178421
  65. There was no statistically significant association between BRAF or MAP2K1 mutation and anatomic site, unifocal versus multifocal presentation, or clinical outcome in Langerhans cell histiocytosis. PMID: 26980021
  66. TNFRSF14 and MAP2K1 mutations are the most frequent genetic alterations found in pediatric-type follicular lymphoma (PTFL) and occur independently in most cases, suggesting that both mutations might play an important role in PTFL lymphomagenesis. PMID: 28533310
  67. MEK1 is constitutively and mainly phosphorylated at the Thr-292, Ser-298, Thr-386, and Thr-388 residues in vivo. Combinations of phosphorylations at these four residues produce at least six phosphorylated variants of MEK1. The phosphorylation statuses of Thr-292, Ser-298, Thr-386, and Thr-388 residues vary widely during activation and deactivation of the MAPK pathway. PMID: 27169363
  68. Combined therapy using a HER2 inhibitor and a BRAF/MEK inhibitor presented a more significant redifferentiation effect on papillary thyroid cancer cells harboring BRAFV600E than BRAF/MEK inhibitor alone. PMID: 28423638
  69. High MEK1 expression is associated with infant acute lymphoblastic leukemia. PMID: 27588400
  70. Mutations in MAP2K1, which are frequently associated with neurological complications and intellectual disability, can be associated with a milder clinical and neurocognitive profile more typical of individuals with Noonan syndrome. Variability of expression may arise from a complex interplay between RAS/MAPK pathway genotype, epigenetics, medical and obstetric factors, and environmental influences. PMID: 27862862
  71. High MEK1 expression is associated with neuroblastoma. PMID: 28687621
  72. There are no other biomarkers correlated with treatment responses following MEK1/2 inhibition. PMID: 27956260
  73. BRAF/MAP2K1-mut LCH cells exhibited a more immature state than BRAF/MAP2K1-wt LCH cells. The BRAFV600E and MAP2K1 mutations were significantly associated with pERK expression. PMID: 27597420
  74. High MEK1 expression is associated with liver cancer. PMID: 26967560
  75. A synthetic lethal interaction of cetuximab in combination with MEK1/2 inhibition for the NRAS mutant subgroup of metastatic colorectal cancer has been reported. PMID: 27636997
  76. MEK1 does not act as a general tumor suppressor in leukemogenesis. Its effects are strongly dependent on the genetic context (RAS versus MYC-driven leukemia) and on the cell type involved. PMID: 27741509
  77. The rs1549854 and rs1432441 polymorphisms of the MAP2K1 gene may be associated with major depressive disorder, especially in females. PMID: 28688265
  78. Mutually exclusive KRAS and MAP2K1 mutations were detected in one-third of cases of Rosai-Dorfman disease. PMID: 28664935
  79. High MEK1 expression is associated with urinary bladder cancer metastasis. PMID: 28534984
  80. Reactive oxygen species-mediated EGFR/MEK/ERK/HIF-1A loop regulates glucose metabolism in pancreatic cancer. PMID: 29702094
  81. Two atypical hairy cell leukemia (HCLc)- and hairy cell leukemia variant (HCLv)-like cases with clinically detected mitogen-activated protein kinase kinase 1 (MAP2K1) mutations have been reported. PMID: 27241017
  82. Activating mutations enhance the rate of MEK1 phosphorylation by Raf. These mutations also affect the thermal stability of MEK1 and its activity towards ERK2. PMID: 29018093
  83. A direct interaction of both MEK1 and MEK2 with AKT has been identified. This interaction between MEK and AKT affects cell migration and adhesion, but not proliferation. The specific mechanism of action of the MEK-AKT complex involves phosphorylation of the migration-related transcription factor FoxO1. PMID: 28225038
  84. Somatic activating mutations in MAP2K1 cause melorheostosis. PMID: 29643386
  85. Upregulation of miR101 inhibited DLBCL cell proliferation and facilitated apoptosis by inhibiting the expression of MEK1. PMID: 30365139
Database Links

HGNC: 6840

OMIM: 176872

KEGG: hsa:5604

STRING: 9606.ENSP00000302486

UniGene: Hs.145442

Involvement In Disease
Cardiofaciocutaneous syndrome 3 (CFC3)
Protein Families
Protein kinase superfamily, STE Ser/Thr protein kinase family, MAP kinase kinase subfamily
Subcellular Location
Cytoplasm, cytoskeleton, microtubule organizing center, centrosome. Cytoplasm, cytoskeleton, microtubule organizing center, spindle pole body. Cytoplasm. Nucleus. Membrane; Peripheral membrane protein.
Tissue Specificity
Widely expressed, with extremely low levels in brain.

Q&A

What is the biological significance of MAP2K1 T292 phosphorylation in MAPK signaling?

T292 phosphorylation on MAP2K1 (also known as MEK1) represents a critical regulatory mechanism within the MAPK/ERK signaling cascade. This phosphorylation event occurs as part of a negative feedback loop where activated ERK phosphorylates MEK1 at T292, which subsequently interferes with PAK-mediated phosphorylation of MEK1 at S298 . This negative feedback mechanism helps maintain homeostatic control of MAPK pathway activation. The T292 phosphorylation acts as a molecular switch that modulates signal duration and intensity by attenuating continued pathway activation. Understanding this phosphorylation event is crucial for interpreting experimental results when studying MAPK pathway dynamics, especially in cancer research where this pathway is frequently dysregulated .

How does the Phospho-MAP2K1 (T292) antibody differ from other MEK1 phospho-antibodies?

The Phospho-MAP2K1 (T292) antibody specifically recognizes MEK1 phosphorylated at threonine 292, distinguishing it from antibodies targeting other phosphorylation sites such as S218/S222 (activation loop phosphorylation) or T386 (another ERK-mediated feedback phosphorylation site) . This specificity allows researchers to selectively monitor the negative feedback regulation of MEK1 by ERK, rather than its activation state. The antibody has been rigorously validated using both wild-type MEK1 and T292A mutant proteins, confirming its specificity for the phosphorylated T292 residue . Unlike antibodies targeting the activation loop phosphorylation (which indicate MEK1 activation), the T292 phospho-antibody provides insight into pathway regulation and signal termination mechanisms, offering complementary information when used alongside other phospho-specific antibodies in signaling studies .

What are the validated applications for Phospho-MAP2K1 (T292) Recombinant Antibody?

The Phospho-MAP2K1 (T292) Recombinant Antibody has been validated for specific research applications through rigorous testing. Current validated applications include:

ApplicationValidation StatusRecommended DilutionNotes
Western Blot (WB)Validated1:1000Successfully detects ~45 kDa phosphorylated MAP2K1
ELISAValidatedVaries by protocolUseful for quantitative analysis

Validation has been performed using recombinant wild-type and mutant (T292A) MAP2K1 proteins, with and without co-expression of MAP kinase, demonstrating specific immunolabeling of the phosphorylated form . The antibody has shown reactivity with human samples and potentially cross-reacts with bovine, dog, mouse, primate, and rat samples due to sequence conservation around the T292 site . It's important to note that optimal dilutions should be determined by each laboratory for specific applications and experimental conditions .

How should controls be designed when using Phospho-MAP2K1 (T292) antibody in Western blot experiments?

Designing appropriate controls is critical for accurately interpreting results with Phospho-MAP2K1 (T292) antibody. A comprehensive control strategy should include:

  • Positive Control: Lysates from cells treated with agents known to induce ERK activation (e.g., EGF, PMA) should show increased T292 phosphorylation due to the negative feedback mechanism .

  • Negative Controls:

    • T292A mutant MEK1 expression constructs (eliminates the phosphorylation site)

    • MEK1 knockdown/knockout cells (validates antibody specificity)

    • MEK-specific inhibitor treatment (e.g., U0126, PD0325901) which should reduce downstream ERK activation and subsequently T292 phosphorylation

  • Loading Controls: Total MEK1 antibody should be used on parallel blots or after stripping to normalize phospho-signal to total protein levels .

  • Pathway Controls: Monitoring ERK activation (phospho-ERK1/2) in parallel is essential since T292 phosphorylation is ERK-dependent .

A technically robust experiment demonstrated in validation studies includes expressing wild-type and T292A mutant MEK1 with and without MAP kinase co-expression, clearly showing that the antibody only detects the phosphorylated form when both wild-type MEK1 and active MAP kinase are present . This approach definitively confirms antibody specificity and is recommended for researchers validating the antibody in their experimental systems.

What cell stimulation conditions optimize detection of MAP2K1 T292 phosphorylation?

Optimal detection of MAP2K1 T292 phosphorylation requires careful consideration of stimulation conditions, as this phosphorylation represents a negative feedback mechanism following ERK activation. Based on the MAPK pathway biology:

  • Temporal Considerations: T292 phosphorylation typically occurs after initial MEK activation and subsequent ERK activation. Time-course experiments are recommended:

    • Short-term stimulation (5-15 minutes): May show initial pathway activation but limited T292 phosphorylation

    • Medium-term stimulation (30-60 minutes): Optimal for detecting T292 phosphorylation as feedback mechanisms engage

    • Long-term stimulation (2+ hours): May show adaptation and reduced signal

  • Effective Stimulants:

    • Growth factors (EGF, PDGF, FGF): 50-100 ng/mL for 30-60 minutes

    • Phorbol esters (PMA): 100-200 nM for 30-60 minutes

    • Serum stimulation: 10-20% FBS for 30-60 minutes after serum starvation

  • Cell Types: Cell lines with robust MAPK pathway activity (e.g., HEK293, NIH3T3, various cancer cell lines) typically show stronger T292 phosphorylation signals .

  • Inhibitor Studies: Using MEK inhibitors (U0126, PD0325901) or ERK inhibitors (SCH772984) can help establish the dependency of T292 phosphorylation on pathway activation. These should eliminate the phospho-signal, confirming specificity .

The detection is optimized when cells are lysed in buffers containing phosphatase inhibitors (sodium fluoride, sodium orthovanadate, and phosphatase inhibitor cocktails) to preserve the phosphorylation status during sample preparation .

What protein extraction methods best preserve MAP2K1 T292 phosphorylation for immunodetection?

Preserving MAP2K1 T292 phosphorylation during protein extraction is critical for accurate detection and quantification. Recommended extraction protocols include:

  • Lysis Buffer Composition:

    • Base buffer: 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% NP-40 or Triton X-100

    • Critical phosphatase inhibitors:

      • 10 mM sodium fluoride

      • 2 mM sodium orthovanadate (freshly activated)

      • 10 mM β-glycerophosphate

      • Commercial phosphatase inhibitor cocktail (1X)

    • Protease inhibitors: PMSF (1 mM) and protease inhibitor cocktail (1X)

  • Extraction Procedure:

    • Perform all steps at 4°C or on ice

    • Scrape cells directly into ice-cold lysis buffer rather than trypsinizing

    • Incubate lysates for 15-20 minutes on ice with occasional vortexing

    • Centrifuge at 14,000 × g for 15 minutes at 4°C

    • Transfer supernatant to fresh tube and avoid freeze-thaw cycles

  • Sample Handling:

    • Add SDS sample buffer immediately to lysates and heat at 95°C for 5 minutes

    • Alternatively, snap-freeze aliquots in liquid nitrogen for long-term storage at -80°C

    • Avoid multiple freeze-thaw cycles which significantly reduce phospho-signal

  • Tissue Samples:

    • Rapidly freeze tissues in liquid nitrogen immediately after collection

    • Pulverize frozen tissue under liquid nitrogen before adding to lysis buffer

    • Homogenize thoroughly while maintaining cold temperature

The effectiveness of phosphorylation preservation can be assessed by comparing phospho-ERK levels in the same samples, as both modifications are phosphatase-sensitive and occur in the same signaling pathway .

How can Phospho-MAP2K1 (T292) antibodies be used to study cross-talk between MAPK and other signaling pathways?

Phospho-MAP2K1 (T292) antibodies provide a valuable tool for investigating the complex cross-talk between MAPK and other signaling pathways. This phosphorylation site serves as a critical node where multiple pathways converge to regulate MAPK signaling:

  • MAPK-PAK Pathway Cross-talk: T292 phosphorylation by ERK interferes with PAK-mediated phosphorylation of MEK1 at S298, creating a phospho-switch mechanism. Researchers can use the T292 phospho-antibody alongside a S298 phospho-antibody to monitor this regulatory cross-talk in real-time following various stimuli .

  • Rac/Cdc42 Signaling Integration: PAK is activated downstream of Rac/Cdc42 small GTPases, so T292 phosphorylation represents a point where Rac/Cdc42 signaling intersects with the MAPK pathway. Experiments combining Rac/Cdc42 activators with MAPK pathway stimulants can reveal how these pathways counter-regulate each other through MEK1 phosphorylation, quantifiable via immunoblotting with the T292 antibody .

  • PI3K-AKT-MAPK Cross-regulation: Multiple studies suggest interactions between PI3K/AKT and MAPK pathways. Researchers can design experiments using specific inhibitors of PI3K (LY294002), AKT (MK2206), and MAPK pathway components while monitoring T292 phosphorylation to map pathway dependencies .

  • Experimental Approach for Cross-talk Studies:

    • Stimulate cells with pathway-specific activators individually and in combination

    • Quantify T292 phosphorylation alongside other pathway-specific phosphorylation events

    • Apply specific pathway inhibitors to define the hierarchy of regulation

    • Monitor temporal dynamics of phosphorylation events to establish causality

This approach has revealed important insights in cancer research, where alterations in feedback mechanisms contribute to therapy resistance and pathway rewiring, particularly in melanoma where MAP2K1/2 mutations occur in approximately 8% of patients .

What are the implications of MAP2K1 T292 phosphorylation in cancer research and therapeutic resistance mechanisms?

MAP2K1 T292 phosphorylation has significant implications in cancer research, particularly regarding therapeutic resistance mechanisms:

Researchers can use the Phospho-MAP2K1 (T292) antibody to assess these mechanisms in patient samples and preclinical models, potentially guiding personalized treatment strategies based on pathway feedback status .

How does MAP2K1 T292 phosphorylation status correlate with cellular responses to targeted MAPK pathway inhibitors?

The phosphorylation status of MAP2K1 at T292 provides critical insights into how cells respond to targeted MAPK pathway inhibitors:

  • Predictive Biomarker for Drug Response: T292 phosphorylation status prior to treatment may indicate the degree of feedback regulation already present in the tumor cells, potentially predicting initial response to MAPK pathway inhibitors. Cells with abnormally low T292 phosphorylation despite high pathway activity may indicate disrupted feedback mechanisms and potentially poorer response to single-agent therapy .

  • Dynamic Biomarker During Treatment: Monitoring T292 phosphorylation during treatment reveals important adaptive responses:

    • Initial decrease in T292 phosphorylation after MEK or RAF inhibitor treatment indicates successful pathway inhibition

    • Rapid recovery of T292 phosphorylation despite continued treatment suggests pathway reactivation and developing resistance

    • Sustained suppression of T292 phosphorylation correlates with durable response

  • Mechanistic Insights from Combination Studies: Research using phospho-specific antibodies has revealed that:

    • MEK inhibitors block downstream ERK activation, reducing T292 phosphorylation

    • RAF inhibitors in BRAF-mutant cells initially decrease T292 phosphorylation

    • In RAS-mutant cells, RAF inhibitors can paradoxically increase T292 phosphorylation due to RAF dimerization

    • Combining RAF and MEK inhibitors more effectively suppresses T292 phosphorylation and improves response durability

  • Correlation with Clinical Outcomes: While more clinical validation is needed, preclinical data suggests that persistent T292 phosphorylation despite MAPK pathway inhibitor treatment correlates with poorer response. Analysis of patient-derived samples before and after treatment progression using Phospho-MAP2K1 (T292) antibodies can provide valuable biomarker data for predicting treatment efficacy .

Researchers can implement regular monitoring of T292 phosphorylation alongside other pathway markers in drug response studies to gain deeper mechanistic understanding of treatment effects and resistance development .

What are common technical challenges when using Phospho-MAP2K1 (T292) antibodies and how can they be resolved?

Researchers commonly encounter several technical challenges when working with Phospho-MAP2K1 (T292) antibodies. Here are evidence-based solutions to these issues:

  • Low Signal Intensity:

    • Cause: Rapid dephosphorylation during sample preparation or low baseline phosphorylation

    • Solution: Enhance phosphatase inhibitor cocktail (double sodium orthovanadate to 4mM); pre-treat cells with phosphatase inhibitors (calyculin A, okadaic acid) for 15-30 minutes before lysis; optimize stimulation conditions to maximize T292 phosphorylation

  • High Background or Non-specific Bands:

    • Cause: Antibody concentration too high; insufficient blocking; cross-reactivity

    • Solution: Optimize antibody dilution (try 1:2000-1:5000); increase blocking time/concentration (5% BSA often works better than milk for phospho-epitopes); add 0.1% Tween-20 to antibody diluent; confirm specificity using T292A mutant as negative control

  • Poor Reproducibility Between Experiments:

    • Cause: Variable phosphorylation status; inconsistent sample preparation

    • Solution: Standardize cell density and stimulation protocol; prepare fresh lysis buffers for each experiment; minimize time between lysis and denaturation; consider using phospho-protein stabilizing buffers commercially available

  • Weak Signal in Tissue Samples:

    • Cause: Delayed processing leading to dephosphorylation; inefficient extraction

    • Solution: Process tissues immediately; snap-freeze in liquid nitrogen; use specialized tissue protein extraction buffers with enhanced phosphatase inhibitors; consider phospho-protein fixation methods prior to extraction

  • Antibody Performance Validation:

    • Problem: Uncertainty about antibody specificity or activity

    • Solution: Always run validation controls (wild-type vs. T292A mutant MEK1 with MAP kinase co-expression) as demonstrated in the technical data ; consider using phosphatase treatment of duplicate samples as additional negative control

The validation methodology shown in multiple sources demonstrates that appropriate controls can distinguish specific signal from artifacts, with clear detection of the ~45 kDa phosphorylated MAP2K1 protein only in samples with active MAP kinase and intact T292 phosphorylation site .

How can multiplexed detection of MAP2K1 phosphorylation sites be optimized for comprehensive pathway analysis?

Optimizing multiplexed detection of different MAP2K1 phosphorylation sites requires careful experimental design to obtain comprehensive pathway analysis. Here's a systematic approach:

  • Antibody Selection and Validation:

    • Choose antibodies raised in different host species (e.g., rabbit anti-phospho-T292, mouse anti-phospho-S218/S222) to enable simultaneous detection

    • Validate each antibody individually using appropriate controls (phosphatase treatment, site-specific mutants)

    • Test for cross-reactivity between antibodies to ensure specific detection

  • Sequential Immunoblotting Approach:

    • Recommended Protocol:

      1. Probe first with anti-phospho-T292 antibody

      2. Document results and strip membrane (validate stripping efficiency)

      3. Reprobe with anti-phospho-S218/S222 antibody

      4. Strip and probe for total MEK1

      5. Strip and probe for loading control (β-actin/GAPDH)

  • Multi-color Fluorescent Western Blotting:

    • Use differentially labeled secondary antibodies (e.g., IRDye 680 and 800)

    • Enables simultaneous detection of two phospho-sites or phospho/total protein ratios

    • Provides more accurate quantification through direct comparison on same blot

  • Sample Preparation Optimization:

    • Different phosphorylation sites may have different sensitivities to phosphatases

    • Use a comprehensive phosphatase inhibitor mixture:

      InhibitorConcentrationTarget Phosphatases
      Sodium fluoride50 mMSerine/threonine phosphatases
      Sodium orthovanadate2-5 mMTyrosine phosphatases
      β-glycerophosphate10 mMSerine/threonine phosphatases
      Sodium pyrophosphate5 mMSerine/threonine phosphatases
      EDTA/EGTA5 mMMetallo-phosphatases
      Commercial cocktail1XBroad spectrum
  • Alternative Technologies for Multiplexed Detection:

    • Phospho-flow cytometry for single-cell resolution

    • Luminex bead-based assays for multiple phospho-proteins

    • Reverse phase protein arrays for high-throughput screening

    • Mass spectrometry for unbiased phospho-site detection

Researchers should note that different phosphorylation sites may have different temporal dynamics after stimulation. T292 phosphorylation (feedback mechanism) typically occurs after activation loop (S218/S222) phosphorylation, so time-course experiments are essential for comprehensive pathway analysis .

How should phospho-specific antibody data be quantified and statistically analyzed for meaningful biological interpretation?

Proper quantification and statistical analysis of phospho-specific antibody data is crucial for deriving meaningful biological interpretations. Here's a comprehensive approach:

  • Quantification Methods:

    • Densitometry Analysis:

      • Use linear range-validated imaging systems (e.g., LI-COR Odyssey, Bio-Rad ChemiDoc)

      • Define regions of interest consistently across all lanes

      • Subtract local background for each lane individually

      • Normalize phospho-signal to total protein rather than housekeeping genes for more accurate comparison

    • Normalization Hierarchy (from most to least accurate):

      1. Phospho-protein/Total target protein ratio from same membrane (multiplex fluorescent detection)

      2. Phospho-protein/Total target protein ratio from stripped and reprobed membrane

      3. Phospho-protein/Total target protein ratio from parallel membranes

      4. Phospho-protein/Loading control ratio (least reliable for phosphorylation studies)

  • Statistical Analysis Framework:

    • For Time-Course Studies:

      • Two-way ANOVA with repeated measures followed by appropriate post-hoc tests

      • Area under the curve (AUC) analysis for comparing sustained vs. transient responses

      • Regression analysis for determining phosphorylation/dephosphorylation rates

    • For Dose-Response Studies:

      • Non-linear regression to determine EC50/IC50 values

      • Calculate fold-change relative to baseline at each concentration

      • Use ANOVA with Dunnett's test to compare treatments to control

    • For Multiple Treatment Comparisons:

      • One-way ANOVA with Tukey's or Bonferroni correction for multiple comparisons

      • Consider mixed-effects models for complex experimental designs

  • Biological Replication Requirements:

    • Minimum three independent biological replicates (different days/passages)

    • Technical replicates cannot substitute for biological replicates

    • Power analysis to determine appropriate sample size (typically n=3-5 for cell studies)

  • Validation Through Orthogonal Methods:

    • Confirm key findings using alternative detection methods:

      • ELISA for quantitative measurement of phospho-T292

      • Immunofluorescence microscopy for spatial information

      • Mass spectrometry for unbiased confirmation

  • Data Presentation Best Practices:

    • Present both representative blots and quantification graphs

    • Include all data points in graphs, not just means and error bars

    • Use box plots or violin plots rather than bar graphs when possible

    • Clearly state statistical tests used and exact p-values

Meaningful interpretation requires correlation with functional outcomes, so researchers should connect phosphorylation data with downstream biological effects and consider pathway modeling approaches for systems-level understanding .

How can MAP2K1 T292 phosphorylation status be utilized in patient stratification for targeted therapy and immunotherapy?

MAP2K1 T292 phosphorylation status represents a potentially valuable biomarker for patient stratification in both targeted therapy and immunotherapy contexts. Current evidence suggests several promising applications:

  • Predictive Biomarker Development:

    • Research has shown that MAP2K1/2 mutations may correlate with response to immune checkpoint inhibitors, particularly CTLA-4 blockade therapy in melanoma patients .

    • T292 phosphorylation status could serve as a functional readout of these mutations' effects on pathway regulation.

    • Immunohistochemistry protocols using phospho-T292 antibodies on patient biopsies could be developed for clinical implementation .

  • Integration with Genetic Biomarkers:

    • Combining T292 phosphorylation analysis with genetic testing for MAP2K1/2 mutations provides complementary information:

      Biomarker CombinationPotential Clinical Interpretation
      MAP2K1/2 mutation (+) / pT292 reducedFunctionally significant mutation disrupting feedback
      MAP2K1/2 mutation (+) / pT292 normalMutation not affecting feedback regulation
      MAP2K1/2 wild-type / pT292 reducedAlternative pathway dysregulation
      MAP2K1/2 wild-type / pT292 normalIntact pathway regulation
  • Therapeutic Decision Support:

    • For targeted therapies: Patients with impaired T292 phosphorylation (disrupted feedback) may benefit from combination treatments that address pathway reactivation

    • For immunotherapy: Based on study data, MAP2K1/2 mutations showed:

      • No significant association with PD-1 inhibitor response (HR = 1.31; 95% CI, 0.68–2.53; p = 0.4151)

      • Possible association with CTLA-4 inhibitor response, suggesting this could be explored as a stratification biomarker

  • Research Implementation Strategy:

    • Develop standardized immunohistochemistry protocols for detecting T292 phosphorylation in FFPE samples

    • Conduct retrospective analysis of T292 phosphorylation in patient cohorts with known treatment outcomes

    • Include T292 phosphorylation analysis in prospective clinical trials as an exploratory endpoint

    • Correlate with other established biomarkers (e.g., TMB, PD-L1 expression)

Future prospective studies should validate the predictive value of combined MAP2K1/2 mutation status and T292 phosphorylation levels to develop clinically applicable stratification algorithms .

What emerging technologies might enhance the detection and functional analysis of MAP2K1 T292 phosphorylation?

Several emerging technologies hold promise for advancing the detection and functional analysis of MAP2K1 T292 phosphorylation with improved sensitivity, specificity, and spatiotemporal resolution:

  • Mass Spectrometry-Based Approaches:

    • Targeted Phosphoproteomics: Selected Reaction Monitoring (SRM) or Parallel Reaction Monitoring (PRM) can quantify T292 phosphopeptides with high sensitivity and specificity

    • Absolute Quantification: Using isotopically labeled synthetic phosphopeptides as internal standards enables absolute quantification of phosphorylation stoichiometry

    • Single-Cell Phosphoproteomics: Emerging techniques for analyzing phosphorylation events at single-cell resolution can reveal heterogeneity in MAP2K1 regulation across cell populations

  • Advanced Imaging Technologies:

    • Super-Resolution Microscopy: Techniques like STORM or PALM combined with phospho-specific antibodies can reveal subcellular localization of phosphorylated MAP2K1

    • Förster Resonance Energy Transfer (FRET): Genetically encoded FRET biosensors for MAP2K1 can monitor T292 phosphorylation dynamics in living cells in real-time

    • Spatial Transcriptomics Integration: Combining phospho-protein detection with spatial transcriptomics can link T292 phosphorylation to local gene expression patterns

  • Functional Genomics Approaches:

    • CRISPR Base Editing: Precise modification of the T292 site to non-phosphorylatable residues without disrupting protein expression

    • Phospho-mimetic Mutations: Expression of T292D/E mutants to model constitutive phosphorylation

    • Optogenetic Control: Light-inducible ERK activation systems to study temporal dynamics of T292 phosphorylation

  • Microfluidic and Lab-on-a-Chip Systems:

    • Phospho-Flow-Seq: Combining phospho-flow cytometry with single-cell RNA sequencing

    • Microfluidic Western Blotting: Miniaturized systems requiring minimal sample input (valuable for limited clinical specimens)

    • Organ-on-a-Chip Models: Testing pathway dynamics in physiologically relevant microenvironments

  • Computational Approaches:

    • Deep Learning Image Analysis: AI-assisted quantification of phospho-T292 immunohistochemistry in tissue samples

    • Dynamic Pathway Modeling: Incorporating T292 phosphorylation kinetics into mathematical models of MAPK pathway behavior

    • Multi-omics Data Integration: Correlating phosphorylation data with transcriptomics, metabolomics and clinical outcomes

These technologies could significantly advance our understanding of MAP2K1 T292 phosphorylation beyond current antibody-based methods, enabling more sophisticated analyses of its role in normal physiology and disease states .

How might therapeutic targeting of the regulatory mechanisms involving MAP2K1 T292 phosphorylation lead to novel cancer treatment strategies?

Targeting the regulatory mechanisms involving MAP2K1 T292 phosphorylation represents an innovative approach to cancer treatment that could address limitations of current MAPK pathway inhibitors:

  • Disrupting Negative Feedback for Enhanced Therapy:

    • Rationale: T292 phosphorylation mediates negative feedback that limits MAPK pathway activation. In certain contexts, enhancing this feedback could suppress oncogenic signaling.

    • Approach: Development of small molecules that mimic or enhance ERK-mediated phosphorylation of T292, potentially stabilizing the phosphorylated state

    • Potential Application: Tumors with hyperactivated MAPK signaling but intact feedback machinery could be sensitive to enhanced negative regulation

  • Targeting Feedback-Escape Mechanisms:

    • Rationale: Some cancers develop resistance by evading T292-mediated feedback inhibition

    • Approaches:

      • Molecules preventing dephosphorylation of T292 by specific phosphatases

      • Compounds that enhance the interaction between phospho-T292 and its effector proteins

      • Peptide mimetics that recapitulate the structural effects of T292 phosphorylation

  • Synergistic Combination Strategies:

    • With MEK Inhibitors: Compounds modulating T292 regulatory mechanisms could enhance durability of response to traditional MEK inhibitors

    • With Immunotherapies: Based on data showing potential correlation between MAP2K1/2 mutations and immunotherapy response, targeting T292 regulatory mechanisms might sensitize tumors to immune checkpoint inhibitors

    • Proposed Combination Strategy:

      Therapeutic ComponentTargetMechanismExpected Outcome
      MEK inhibitorMEK catalytic activityBlock downstream signalingInitial pathway inhibition
      T292 regulatory modulatorFeedback mechanismPrevent pathway reactivationPrevent resistance development
      Immunotherapy (CTLA-4 inhibitor)Immune checkpointEnhance anti-tumor immunitySustained tumor control
  • Patient Selection Strategies:

    • Studies indicate MAP2K1/2 mutations may predict response to CTLA-4 inhibitors but not PD-1 inhibitors in melanoma

    • Therapies targeting T292 regulatory mechanisms could be most effective in patients with:

      • Wild-type MAP2K1/2 with intact feedback regulation

      • Specific MAP2K1/2 mutations that alter but don't eliminate T292 phosphorylation

      • Tumors showing primary or acquired resistance to conventional MAPK inhibitors

  • Therapeutic Monitoring Approach:

    • Serial biopsies to track T292 phosphorylation status during treatment

    • Development of surrogate biomarkers (e.g., plasma phospho-proteomics)

    • Integration with imaging technologies to assess pathway modulation in vivo

This targeted approach to pathway regulation, rather than simple inhibition, represents a paradigm shift in MAPK pathway-directed therapies that could potentially overcome current limitations in treatment durability and response .

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.