Phospho-IGF1R (Tyr1165/Tyr1166) Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Phospho-IGF1R (Tyr1165/Tyr1166) Antibody

Phospho-IGF1R (Tyr1165/Tyr1166) Antibody is a specialized immunological tool designed to recognize and bind specifically to IGF1R protein only when phosphorylated at tyrosine residues 1165 and 1166. This antibody is predominantly produced in rabbits as polyclonal antibodies, generated by immunizing the animals with synthetic phosphopeptides corresponding to regions surrounding these specific phosphorylation sites in human IGF1R. The resulting antibodies are then purified through affinity chromatography using the immunizing phosphopeptide to ensure high specificity .

The development of phospho-specific antibodies like Phospho-IGF1R (Tyr1165/Tyr1166) Antibody has revolutionized signal transduction research by enabling scientists to monitor the activation status of key signaling molecules. These antibodies have become invaluable for investigating the complex regulatory mechanisms controlling IGF1R activation and its downstream signaling cascades that influence cellular proliferation, survival, and metabolism .

The high specificity of these antibodies permits researchers to differentiate between the active (phosphorylated) and inactive forms of IGF1R, providing crucial insights into cellular signaling events that depend on IGF1R activation. This specificity is essential for understanding the role of IGF1R in various biological contexts, particularly in cancer research where IGF1R signaling is frequently dysregulated .

Structure and Biological Significance of IGF1R

IGF1R is a transmembrane receptor tyrosine kinase that plays critical roles in development, cell growth, and metabolism. The receptor exists as a tetramer composed of two alpha and two beta subunits. The alpha subunits are located extracellularly and are responsible for binding to insulin-like growth factors, while the beta subunits contain the tyrosine kinase domain responsible for signal transduction .

The receptor binds insulin-like growth factor 1 (IGF1) with high affinity and insulin-like growth factor 2 (IGF2) with lower affinity. This binding triggers a conformational change in the receptor, followed by autophosphorylation at multiple tyrosine residues, including positions 1165 and 1166 . This autophosphorylation activates the intrinsic tyrosine kinase activity of IGF1R, enabling it to phosphorylate substrate proteins and initiate downstream signaling cascades.

The activated IGF1R initiates several major signaling pathways, including:

  1. The RAS/RAF/MAPK pathway, which promotes cell proliferation

  2. The PI3K/AKT pathway, which enhances cell survival and protein synthesis

  3. The JAK/STAT pathway, which regulates gene transcription

These pathways collectively contribute to diverse cellular responses, including proliferation, differentiation, survival, and metabolism . Dysregulation of IGF1R signaling has been implicated in various pathological conditions, most notably in cancer, where it contributes to tumor transformation and the survival of malignant cells .

Phosphorylation Mechanism and Significance of Tyr1165/Tyr1166

The phosphorylation of tyrosine residues 1165 and 1166 is a critical event in the activation of IGF1R. These residues are located within the catalytic loop of the tyrosine kinase domain and their phosphorylation is essential for the full activation of the receptor's kinase activity .

Autophosphorylation of IGF1R occurs in a sequential and trans manner, meaning that one subunit of the dimeric receptor phosphorylates tyrosine residues on the other subunit. Tyr-1165 is predominantly phosphorylated first, followed by phosphorylation of Tyr-1166. While each individual phosphorylation increases kinase activity, optimal activation requires phosphorylation of multiple tyrosine residues in the kinase activation loop .

The catalytic loops within the tyrosine kinase domains of IGF1R contain a three-tyrosine motif corresponding to Tyr1131, Tyr1135, and Tyr1136. This phosphorylation pattern is remarkably similar to the insulin receptor (IR), which contains corresponding residues at positions Tyr1150 and Tyr1151. Due to this structural similarity, some Phospho-IGF1R (Tyr1165/Tyr1166) antibodies may cross-react with the phosphorylated insulin receptor .

The phosphorylation status of Tyr1165/Tyr1166 serves as a biomarker for IGF1R activation and provides valuable information about the activity of IGF1R-dependent signaling pathways in various cellular contexts, particularly in cancer cells where IGF1R signaling is often upregulated .

Research Applications of Phospho-IGF1R (Tyr1165/Tyr1166) Antibody

Phospho-IGF1R (Tyr1165/Tyr1166) Antibody serves as a valuable tool in various research applications aimed at understanding IGF1R signaling and its implications in health and disease. The primary applications include:

Western Blotting (WB)

Western blotting represents one of the most common applications for this antibody. This technique allows researchers to detect and semi-quantify the levels of phosphorylated IGF1R in cell or tissue lysates. The antibody typically detects bands at approximately 95-155 kDa, corresponding to the phosphorylated beta subunit of IGF1R . Western blotting is particularly useful for studying the activation status of IGF1R in response to various stimuli, such as treatment with insulin or IGF1, or in different disease models.

Enzyme-Linked Immunosorbent Assay (ELISA)

ELISA provides a quantitative measure of phosphorylated IGF1R levels in samples. With typical recommended dilutions ranging from 1:1000 to 1:20000, this application offers high sensitivity for detecting even small changes in phosphorylation status . ELISA is useful for high-throughput screening of compounds that may affect IGF1R phosphorylation and for comparing phosphorylation levels across multiple samples or conditions.

Immunohistochemistry (IHC) and Immunocytochemistry (ICC)

Immunohistochemistry applications allow for the visualization of phosphorylated IGF1R within tissue sections, while immunocytochemistry enables detection in cultured cells. These techniques provide valuable information about the spatial distribution of activated IGF1R . In cancer research, IHC is particularly useful for assessing the activation status of IGF1R in tumor samples, which may correlate with disease progression or response to therapy.

Validation Methods

To ensure the specificity of Phospho-IGF1R (Tyr1165/Tyr1166) Antibody, various validation methods are employed:

  1. Peptide competition assays: The antibody is pre-incubated with the immunizing phosphopeptide before application, which should block specific binding and eliminate signal

  2. Non-phosphorylated peptide controls: Pre-incubation with non-phosphorylated peptide should not affect antibody binding

  3. Cell stimulation experiments: Cells treated with IGF1 or insulin should show increased phospho-IGF1R signal compared to untreated cells

  4. Phosphatase treatment: Samples treated with phosphatases should show reduced or eliminated signal

GeneTex's product has demonstrated high specificity through peptide competition assays, showing that only the immunogen phosphopeptide blocks the signal, while non-phosphorylated peptide and generic phosphotyrosine-containing peptide controls do not affect antibody binding .

IGF1R Signaling in Cancer and Disease

IGF1R signaling has been implicated in various diseases, with particularly strong evidence for its role in cancer development and progression. Understanding the phosphorylation status of IGF1R at Tyr1165/Tyr1166 provides valuable insights into disease mechanisms and potential therapeutic targets.

Role in Cancer Development

IGF1R is highly expressed in most malignant tissues, where it functions as an anti-apoptotic agent by enhancing cell survival . The receptor is crucial for tumor transformation and the survival of malignant cells. Upon ligand binding and phosphorylation at Tyr1165/Tyr1166, IGF1R activates downstream signaling pathways that promote:

  1. Increased cellular proliferation through the MAPK pathway

  2. Inhibition of apoptosis through the PI3K-AKT/PKB pathway

  3. Enhanced protein synthesis through mTOR activation

These mechanisms collectively contribute to cancer cell survival, proliferation, and resistance to therapy . The phosphorylation status of IGF1R at Tyr1165/Tyr1166 serves as a biomarker for receptor activation and may correlate with disease progression or response to targeted therapies.

Physiological and Pathological Significance

Beyond cancer, IGF1R signaling plays crucial roles in normal development and metabolism. The receptor is involved in:

  1. Embryonic growth and development

  2. Tissue regeneration and repair

  3. Regulation of glucose metabolism

  4. Neuronal survival and function

Dysregulation of IGF1R signaling has been implicated in various pathological conditions, including:

  1. Metabolic disorders such as diabetes and obesity

  2. Neurodegenerative diseases

  3. Aging-related conditions

  4. Cardiovascular diseases

The ability to detect and quantify phosphorylated IGF1R at Tyr1165/Tyr1166 using specific antibodies provides researchers with a valuable tool for investigating the role of this signaling pathway in both physiological and pathological contexts .

Experimental Considerations and Protocols

When working with Phospho-IGF1R (Tyr1165/Tyr1166) Antibody, several experimental considerations should be addressed to ensure reliable and reproducible results:

Sample Preparation

Proper sample preparation is crucial for detecting phosphorylated IGF1R. Key considerations include:

  1. Including phosphatase inhibitors in lysis buffers to preserve phosphorylation status

  2. Processing samples quickly to minimize dephosphorylation

  3. Using appropriate positive controls, such as cells stimulated with IGF1 or insulin

  4. Including negative controls, such as unstimulated cells or phosphatase-treated samples

Protocol Optimization

For optimal results with Phospho-IGF1R (Tyr1165/Tyr1166) Antibody, protocol optimization may include:

  1. Determining the optimal antibody dilution for each application (typical ranges: WB 1:500-1:2000, ELISA 1:1000-1:20000, IHC 1:50-1:100)

  2. Optimizing blocking conditions to minimize background signal

  3. Adjusting incubation times and temperatures for optimal signal-to-noise ratio

  4. Implementing appropriate washing steps to remove unbound antibody

Cross-Reactivity Considerations

Due to the sequence similarity between IGF1R and the insulin receptor (IR), some Phospho-IGF1R (Tyr1165/Tyr1166) Antibodies may cross-react with phosphorylated IR at the corresponding sites (Tyr1150/Tyr1151) . This cross-reactivity should be considered when interpreting results, especially in tissues or cells that express both receptors. When specific detection of phosphorylated IGF1R without IR cross-reactivity is required, additional validation experiments or alternative antibodies may be necessary.

Recent Advances and Future Directions

Recent advances in antibody technology have led to improved Phospho-IGF1R (Tyr1165/Tyr1166) Antibodies with enhanced specificity and sensitivity. These improvements have facilitated more detailed investigations of IGF1R signaling in various biological contexts.

Emerging Applications

Emerging applications for Phospho-IGF1R (Tyr1165/Tyr1166) Antibody include:

  1. Single-cell analysis of IGF1R activation in heterogeneous cell populations

  2. In vivo imaging of IGF1R activation in animal models

  3. Companion diagnostics for IGF1R-targeted therapies

  4. High-throughput screening for compounds that modulate IGF1R phosphorylation

Future Directions

Future directions in this field may include:

  1. Development of monoclonal antibodies with even higher specificity for phosphorylated IGF1R

  2. Creation of antibodies that can distinguish between different phosphorylation patterns on IGF1R

  3. Integration of phospho-IGF1R detection with other biomarkers for more comprehensive signaling analysis

  4. Application of these antibodies in precision medicine approaches to stratify patients for IGF1R-targeted therapies

These advancements will continue to enhance our understanding of IGF1R signaling and its implications in health and disease.

Product Specs

Form
Supplied at 1.0 mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150 mM NaCl, 0.02% sodium azide, and 50% glycerol.
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your orders. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery time estimates.
Synonyms
CD221 antibody; CD221 antigen antibody; IGF 1 receptor antibody; IGF 1R antibody; IGF I receptor antibody; IGF-I receptor antibody; Igf1r antibody; IGF1R_HUMAN antibody; IGFIR antibody; IGFIRC antibody; IGFR antibody; Insulin like growth factor 1 receptor antibody; Insulin like growth factor 1 receptor precursor antibody; Insulin-like growth factor 1 receptor beta chain antibody; Insulin-like growth factor I receptor antibody; JTK13 antibody; MGC142170 antibody; MGC142172 antibody; MGC18216 antibody; Soluble IGF1R variant 1 antibody; Soluble IGF1R variant 2 antibody
Target Names
Uniprot No.

Target Background

Function
The insulin-like growth factor 1 receptor (IGF1R) is a receptor tyrosine kinase that mediates the actions of insulin-like growth factor 1 (IGF1). It binds IGF1 with high affinity and IGF2 and insulin (INS) with lower affinity. The activated IGF1R plays a vital role in cell growth and survival control. IGF1R is crucial for tumor transformation and survival of malignant cells. Upon ligand binding, the receptor kinase is activated, leading to receptor autophosphorylation and tyrosine phosphorylation of multiple substrates that function as signaling adapter proteins. These substrates include the insulin-receptor substrates (IRS1/2), Shc, and 14-3-3 proteins. Phosphorylation of IRS proteins activates two major signaling pathways: the PI3K-AKT/PKB pathway and the Ras-MAPK pathway. The MAPK pathway activation results in increased cellular proliferation, while activation of the PI3K pathway inhibits apoptosis and stimulates protein synthesis. Phosphorylated IRS1 can activate the 85 kDa regulatory subunit of PI3K (PIK3R1), leading to the activation of several downstream substrates, including protein AKT/PKB. AKT phosphorylation, in turn, enhances protein synthesis through mTOR activation and triggers the antiapoptotic effects of IGFIR through phosphorylation and inactivation of BAD. In parallel to PI3K-driven signaling, the recruitment of Grb2/SOS by phosphorylated IRS1 or Shc leads to the recruitment of Ras and activation of the ras-MAPK pathway. In addition to these two main signaling pathways, IGF1R also signals through the Janus kinase/signal transducer and activator of transcription pathway (JAK/STAT). Phosphorylation of JAK proteins can lead to phosphorylation/activation of signal transducers and activators of transcription (STAT) proteins. In particular, activation of STAT3 may be essential for the transforming activity of IGF1R. The JAK/STAT pathway activates gene transcription and may be responsible for the transforming activity. JNK kinases can also be activated by the IGF1R. IGF1 exerts inhibiting activities on JNK activation via phosphorylation and inhibition of MAP3K5/ASK1, which is able to directly associate with the IGF1R. When present in a hybrid receptor with INSR, it binds IGF1. PubMed:12138094 demonstrates that hybrid receptors composed of IGF1R and INSR isoform Long are activated with high affinity by IGF1, with low affinity by IGF2 and not significantly activated by insulin. It also shows that hybrid receptors composed of IGF1R and INSR isoform Short are activated by IGF1, IGF2, and insulin. In contrast, PubMed:16831875 shows that hybrid receptors composed of IGF1R and INSR isoform Long and hybrid receptors composed of IGF1R and INSR isoform Short have similar binding characteristics, both bind IGF1 and have a low affinity for insulin.
Gene References Into Functions
  1. MiR133a and miR133b may bind near rs1815009, and miR455 near rs2684788, within IGF1R 3'UTR. PMID: 30365147
  2. This study confirms the utility of proximity-labeling methods, such as BioID, to screen for interactors of cell-surface receptors and has uncovered a role of one of these interactors, SNX6, in the IGF1R signaling cascade. PMID: 29530981
  3. Elevations of TGF-beta3, SMAD2, and SMAD4 in hypertrophic scars and an increase of IGF-1R in immature stages may provide insights into acne hypertrophic scar formation. PMID: 30167815
  4. miR-30a-5p could influence chemo-resistance by targeting IGF1R gene in melanoma cells, potentially providing a target for the therapy of chemo-resistant melanoma cells. PMID: 29642855
  5. IGF-1R signaling contributes to T cell-dependent inflammation in arthritis. Inhibition of IGF-1R at the level of insulin receptor substrates alleviates arthritis by restricting IL6-dependent formation of Th17 cells and may open up new treatment strategies for rheumatoid arthritis. PMID: 28583713
  6. A novel G310D variant in the insulin-like growth factor 1 receptor gene is associated with type 2 diabetes. PMID: 29470850
  7. Higher IGF-IR mRNA expression observed in obese children is associated with higher IGF-I and ALS and lower IGFBP-1 levels. PMID: 29150385
  8. Study results indicate that microRNA-320a suppresses tumor cell growth and invasion of human breast cancer by targeting IGF-1R. PMID: 29989645
  9. miR539 may inhibit the aggressive behavior of PDAC by directly targeting IGF1R and may serve as a novel therapeutic target for patients with this disease. PMID: 29901181
  10. Data suggest that NEAT1, SRC3, and IGF1R are highly expressed in prostate cancer cells; NEAT1 appears to interact with SRC3 and promote cell proliferation via up-regulation of the SRC3/IGF1R/AKT signaling pathway. (NEAT1 = nuclear paraspeckle assembly transcript-1; SRC3 = steroid receptor coactivator protein-3; IGF1R = insulin-like growth factor 1 receptor) PMID: 29225160
  11. Results showed that the expression of IGF1R appears to be highly correlated with the expression of ABCG2 in osteosarcoma and with the expression of CD44 in osteosarcoma patients under the age of 10. PMID: 29892839
  12. This study reports a nodal role of IGF-IR in the regulation of ERalpha-positive breast cancer cell aggressiveness and the regulation of expression levels of several extracellular matrix molecules. PMID: 28079144
  13. Long noncoding RNA PVT1 enhances the expression of IGF1R through competitive binding to miR-30a. PMID: 29803929
  14. The association between the growth hormone-insulin-like growth factor-1 (GH-IGF-1) axis gene polymorphisms and short stature in Chinese children. PMID: 29687007
  15. High IGF1R expression is associated with non-small cell lung cancer. PMID: 29328495
  16. Our findings suggest that CKS1BP7 as well as IGF1R may serve as potential biomarkers for early detection and predict prognosis in breast cancer. PMID: 28439706
  17. High IGF-IR expression is associated with Ras and BRAF mutations in Hepatocellular Carcinoma. PMID: 28188432
  18. MicroRNA-381 inhibits cell proliferation and invasion in endometrial carcinoma by targeting the IGF-1R. PMID: 29257334
  19. IGF-1R and AKT inhibitors further increased apoptosis by Nutlin-3a in parental MHM cells and the cisplatin-resistant clones, confirming that IGF-1R/AKT signaling promotes apoptosis resistance. PMID: 28696156
  20. Autocrine IGF2 constitutively activated IGF1R and Akt phosphorylation, which was inhibited by BI 885578 treatment. BI 885578 significantly delayed the growth of IGF2-high colorectal cancer xenograft tumors in mice, while combination with a VEGF-A antibody increased efficacy and induced tumor regression. PMID: 28729397
  21. These findings demonstrated that hMSCCMmediated neuroprotection was attributed to IGF1Rmediated signaling, potentiated via the inhibition of IGF2 by IGFBP6. The results of this study provide insight into the mechanism by which hMSC administration may promote recovery from nerve injury. PMID: 29039467
  22. Loss of miR-99a in ESCC promoted tumor cell proliferation, migration, invasion, and slug-induced EMT through activating the IGF1R signaling pathway. PMID: 28800315
  23. Current data demonstrate that both INSR and IGF1R are directly targeted by C-myc and exert similar effects to promote the tumorigenesis and metastasis of TSCC through the NF-kappaB pathway. PMID: 29518496
  24. WP760 downregulated IGF1R. PMID: 28417283
  25. This study shows a cross-talk between IGF1R and Wnt/beta-catenin signaling pathways and demonstrates, for the first time, that IGF1R is associated with upregulation of TCF-mediated beta-catenin transcriptional activity. PMID: 29621572
  26. MicroRNA-Dependent Regulation of IGF1R Gene Expression in Hormone-Sensitive and Hormone-Resistant Prostate Cancer Cells PMID: 29779108
  27. In contrast to preclinical studies that suggest a decrease in trastuzumab sensitivity in IGF1R(+) tumors, our adjuvant data show benefit of adding trastuzumab for patients with either IGF1R(+) and IGF1R(-) breast tumors. PMID: 28348046
  28. The findings demonstrate that miR-186 acts as a tumor suppressor by targeting IGF-1R in glioma. PMID: 28944896
  29. Forced expression of Klotho resulted in a decline of activation of IGF-1R signaling, accompanied by decreased phosphorylation of its downstream targets, including AKT and ERK1/2. These data indicated that Klotho acts as a tumor suppressor via inhibiting IGF-1R signaling, thus suppressing viability and promoting apoptosis in T-cell lymphoma. PMID: 28656297
  30. The study concluded that the expression modulation of tumor suppressors MIR-375 and MIR-145, and oncomiR MIR-224 have the ability to induce apoptosis of colorectal carcinoma cells through regulation of apoptosis mediating genes MTDH, MAP3K1, PDK1, BCL-XL, and BAX. PMID: 28802228
  31. Activation of the IGF-IR/PI3K/Akt signaling system is a common pattern in MLS, which appears to be transcriptionally controlled, at least in part by induction of IGF2 gene transcription in a FUS-DDIT3-dependent manner. PMID: 28637688
  32. Implantation of IGF1R(+) human dental pulp mesenchymal stem cells exerted enhanced neuroplasticity via integrating inputs from both CXCR4 and IGF1R signaling pathways. PMID: 27586516
  33. Study findings indicate that the T allele of IGF1R variant rs2016347 is associated with a significant reduction in breast cancer risk in women with a history of preeclampsia, most marked for HR+ breast cancer and in women with age at first birth less than 30. PMID: 28822014
  34. This study suggests that IGF-1R-AKT signaling imparts functional heterogeneity in cancer stem cells during the acquirement of chemoresistance in ovarian carcinoma. PMID: 27819360
  35. IGF1R mRNA expression levels were reversely correlated with miR503 expression levels in breast tumors, suggesting that the upregulation of IGF1R may be due to downregulation of miR503 in breast cancer. PMID: 28656281
  36. miR-497 and miR-99a synergistically target IGF1R and mTOR, thereby impeding HCC tumor growth. These results promote a concept in which not one single miRNA, but rather a network of miRNAs with shared and individual mRNA targets participates in hepatocarcinogenesis. PMID: 28624790
  37. MiR379 acts as a tumor suppressor in NSCLC by directly targeting IGF1R. PMID: 28731178
  38. These results indicate that miR455 is involved in gastric cancer progression by directly targeting IGF1R and may serve as a novel therapeutic target for the treatment of gastric cancer. PMID: 28714005
  39. Tumor cells in CSF express IGF1R in High Risk, Metastatic Medulloblastoma. PMID: 27255663
  40. Insulin-like growth factor 1 receptor, associate of Myc 1, and peroxisome proliferator-activated receptor gamma coactivator 1beta are direct targets of miR-139. PMID: 26868851
  41. THADA fusion is a mechanism of IGF2BP3 activation and IGF1R signaling in thyroid cancer. PMID: 28193878
  42. In addition to conventional methods, IGF1R CNV can be identified from WES data. FACS analysis of live primary cells is a promising method for efficiently evaluating and screening for IGF1R haploinsufficiency. PMID: 28395282
  43. The T IGFR-1 genetic variant and a combination of the C VEGF-A and T IGFR-1 genetic variants increase the risk of developing Primary Open Angle Glaucoma. PMID: 28745651
  44. In endocrine-sensitive breast cancer cells, insulin was not growth stimulatory, likely due to the presence of hybrid InsR/IGF1R, which has high affinity for IGF-I but not insulin. Combination inhibition of InsR and IGF1R showed complete suppression of the system in endocrine-sensitive breast cancer cells. PMID: 28468775
  45. Lower IGF-1R expression after teriparatide was associated with higher body fat, suggesting links between teriparatide resistance, body composition, and the GH/IGF-1 axis. PMID: 28218468
  46. This study showed that IGF-1 receptor (IGF-1R), which mediates survival pathways upon IGF binding, was highly expressed in oculomotor neurons and on extraocular muscle endplate. PMID: 27180807
  47. This study confirmed the tumor suppressor function of miR-455 in melanoma and demonstrated that miR-455 suppressed proliferation and invasion through directly targeting IGF-1R. PMID: 28440508
  48. IGF1R signaling under the given experimental conditions and NSCLC genetic background dictates the functional endpoint mechanism for TKI resistance. Manipulating this regulatory role of IGF1R can force the functional endpoint mechanism for TKI resistance in a defined and targetable direction, as illustrated by the observed MET-amplification. PMID: 28418902
  49. This report describes complex relationships between individual tumor-specific expression of IGF1R/pIGF1R and InsR/pInsR, response to endocrine treatment, and breast cancer prognosis. PMID: 28030849
  50. These data imply the potential clinical application of EGF-LDP-IGF-AE for esophageal squamous cell carcinoma (ESCC) patients with EGFR and/or IGF-1R overexpression. PMID: 28498434

Show More

Hide All

Database Links

HGNC: 5465

OMIM: 147370

KEGG: hsa:3480

STRING: 9606.ENSP00000268035

UniGene: Hs.643120

Involvement In Disease
Insulin-like growth factor 1 resistance (IGF1RES)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, Insulin receptor subfamily
Subcellular Location
Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
Found as a hybrid receptor with INSR in muscle, heart, kidney, adipose tissue, skeletal muscle, hepatoma, fibroblasts, spleen and placenta (at protein level). Expressed in a variety of tissues. Overexpressed in tumors, including melanomas, cancers of the

Q&A

What is the biological significance of IGF1R phosphorylation at Tyr1165/Tyr1166 sites?

The phosphorylation of IGF1R at Tyr1165/Tyr1166 represents a critical activation event in the IGF1R signaling pathway. These tyrosine residues are located within the activation loop of the receptor's kinase domain. When IGF-1 or IGF-2 binds to the alpha subunits of the receptor, it triggers a conformational change that leads to autophosphorylation of the receptor. Phosphorylation occurs in a sequential manner, with Tyr1165 being predominantly phosphorylated first, followed by phosphorylation of Tyr1166 .

This specific phosphorylation event is essential for full activation of the receptor's intrinsic tyrosine kinase activity. Once phosphorylated, IGF1R can then phosphorylate downstream substrate proteins, initiating signaling cascades that involve sequential activation of RAS, RAF, and mitogen-activated protein kinase isoforms (ERK, p38, and JNK). These pathways ultimately lead to the transcription of genes that drive cellular proliferation, differentiation, and survival .

How does the phosphorylation sequence of IGF1R differ from the insulin receptor, and why is this important?

IGF1R and the insulin receptor (IR) share significant structural homology and overlapping signaling mechanisms. In the insulin receptor, the catalytic loop contains tyrosine residues numbered as Tyr1158, Tyr1162, and Tyr1163 (according to Ebina et al.) or Tyr1150 and Tyr1151 (according to Ullrich et al.). The corresponding residues in IGF1R are Tyr1131, Tyr1135, and Tyr1136, with Tyr1165 and Tyr1166 being alternative numbering systems for some of these sites .

Autophosphorylation of both receptors proceeds in a processive manner, initiating at the second tyrosine (Tyr1162 in IR; Tyr1135 in IGF1R), followed by phosphorylation at the other tyrosines, resulting in full activation of the receptors . Despite these similarities, each receptor has unique phosphorylation sites that presumably contribute to the distinct signaling outcomes between insulin and IGF pathways .

Understanding these differences is crucial for designing specific antibodies that can discriminate between activated IGF1R and IR, particularly in research focused on metabolic disorders or cancer where these pathways may be differentially regulated .

What criteria should be considered when selecting a Phospho-IGF1R (Tyr1165/Tyr1166) antibody for specific applications?

When selecting a Phospho-IGF1R (Tyr1165/Tyr1166) antibody, researchers should consider several critical factors:

  • Specificity for phosphorylated sites: Ensure the antibody has been validated to specifically detect IGF1R only when phosphorylated at Tyr1165/Tyr1166, as demonstrated through phosphopeptide competition assays .

  • Cross-reactivity with insulin receptor: Due to sequence homology, determine whether the antibody cross-reacts with the equivalent phosphorylated sites on the insulin receptor. Some antibodies are designed to detect both receptors, while others are IGF1R-specific .

  • Species reactivity: Confirm the antibody recognizes the target across relevant experimental species (human, mouse, rat, etc.) .

  • Validated applications: Verify that the antibody has been validated for your specific application (Western blot, immunocytochemistry, ELISA, etc.) .

  • Clonality: Consider whether a polyclonal or monoclonal antibody is more suitable for your research needs. Polyclonal antibodies may offer higher sensitivity but potentially lower specificity .

  • Sensitivity: Evaluate the antibody's ability to detect low levels of phosphorylated receptor, especially important in studies involving subtle changes in activation status .

  • Background signals: Assess reports of non-specific binding or background noise in similar experimental contexts .

The search results indicate that most commercial Phospho-IGF1R (Tyr1165/Tyr1166) antibodies are rabbit polyclonal antibodies validated for applications such as Western blot, ELISA, and immunocytochemistry across human, mouse, and rat samples .

What are the differences between antibodies recognizing dual phosphorylation (Tyr1165/Tyr1166) versus triple phosphorylation sites of IGF1R?

Antibodies targeting dual phosphorylation sites (Tyr1165/Tyr1166) differ significantly from those recognizing triple phosphorylation sites (Tyr1131/Tyr1135/Tyr1136 or Tyr1161/Tyr1165/Tyr1166) in several important aspects:

Dual phosphorylation antibodies:

  • Recognize IGF1R when phosphorylated specifically at Tyr1165 and Tyr1166

  • May detect intermediate activation states of the receptor

  • Typically generated using a synthetic phosphopeptide with the sequence motif T-D-Y(p)-Y(p)-R-K

  • Often used to detect early activation events in the IGF1R signaling cascade

Triple phosphorylation antibodies:

  • Recognize IGF1R when phosphorylated at all three key tyrosine residues in the activation loop

  • Generally indicate fully activated receptor status

  • May provide higher stringency for detecting completely activated receptors

  • Often generated using immunogens containing all three phosphorylated tyrosines

The choice between these antibodies depends on research objectives. Dual phosphorylation antibodies might be preferred for detecting initial activation events, while triple phosphorylation antibodies might better indicate maximal kinase activity. Studies by Patel et al. demonstrated that while every single phosphorylation increases kinase activity, all three tyrosine residues in the kinase activation loop have to be phosphorylated for optimal activity .

What are the optimal conditions for detecting phospho-IGF1R (Tyr1165/Tyr1166) in Western blot applications?

Optimizing Western blot detection of phospho-IGF1R (Tyr1165/Tyr1166) requires careful attention to multiple experimental parameters:

Sample preparation:

  • Rapidly harvest cells to preserve phosphorylation status

  • Use phosphatase inhibitors in lysis buffers to prevent dephosphorylation

  • Standardize protein loading (30-50 μg total protein per lane is typically sufficient)

  • Include positive controls such as insulin/IGF-1 stimulated cell lysates (e.g., 293 cells or CHO-T cells treated with 100 nM insulin/IGF-1 for 5-10 minutes)

Experimental conditions:

  • Dilution range: Most antibodies work optimally at 1:500-1:2000 dilution for Western blot

  • Blocking: 5% BSA in TBST is generally preferred over milk for phospho-specific antibodies

  • Primary antibody incubation: Overnight at 4°C for optimal signal-to-noise ratio

  • Secondary antibody: Use high-quality HRP-conjugated anti-rabbit IgG (typically at 1:5000-1:10000)

Signal detection:

  • Enhanced chemiluminescence (ECL) systems with varying sensitivity options based on expected expression levels

  • Stripping and reprobing membranes with total IGF1R antibodies for normalization

  • Quantification using appropriate software and normalizing phospho-signal to total receptor levels

Troubleshooting:

  • High background: Increase antibody dilution or washing steps

  • Weak signal: Enrich for membrane proteins or immunoprecipitate receptor before Western blot

  • Multiple bands: Validate specificity using phosphopeptide competition assays as demonstrated in search result

For validation, peptide competition assays can be performed by pre-incubating the antibody with phosphopeptide immunogen, non-phosphorylated peptide, or generic phosphotyrosine-containing peptide before probing the membrane .

How can researchers effectively use phospho-IGF1R (Tyr1165/Tyr1166) antibodies in cell-based ELISA assays?

Cell-based ELISA assays provide quantitative determination of phospho-IGF1R (Tyr1165/Tyr1166) expression directly in cultured cells. For optimal results, researchers should follow these methodological guidelines:

Assay setup:

  • Seed cells at consistent density in 96-well plates (typically 1-3 × 10^4 cells per well)

  • Include both stimulated (IGF-1/insulin treated) and unstimulated control wells

  • Fix cells with 4% paraformaldehyde after treatment to preserve phosphorylation state

  • Permeabilize with 0.1-0.5% Triton X-100 to allow antibody access to intracellular epitopes

Antibody incubation:

  • Use phospho-IGF1R (Tyr1165/Tyr1166) antibodies at manufacturer-recommended dilutions (typically 1:100-1:500)

  • Include appropriate controls: primary antibody omission, isotype controls, and phosphatase-treated samples

  • For dual readout assays, include anti-GAPDH or anti-total IGF1R antibodies for normalization

Normalization methods:
Based on search result , multiple normalization approaches are recommended:

  • Anti-GAPDH antibody as an internal positive control

  • Crystal Violet whole-cell staining for cell density normalization

  • Anti-total IGF1R antibody for normalizing to receptor expression levels

Data analysis:

  • Calculate phospho-IGF1R/total IGF1R ratios to account for variations in receptor expression

  • For multi-condition experiments, express results as fold-change relative to unstimulated controls

  • Statistical analysis should account for biological replicates across multiple independent experiments

This approach allows for qualitative determination of IGF1R phosphorylation status and comparative analysis of how different stimulation conditions affect receptor activation in various cell lines .

What cell types and stimulation protocols are most appropriate for studying IGF1R phosphorylation at Tyr1165/Tyr1166?

Selecting appropriate cell models and stimulation protocols is crucial for investigating IGF1R phosphorylation at Tyr1165/Tyr1166. Based on the search results, the following approaches have proven effective:

Recommended cell types:

  • MCF-7 cells: Human breast cancer cell line with well-characterized IGF1R expression, recommended as a positive control for ICC/IF studies

  • 293 (HEK293) cells: Commonly used for overexpression studies and as positive controls for Western blot

  • CHO-T cells: Chinese hamster ovary cells transfected with human insulin receptor, useful for cross-reactivity studies

  • 3T3-L1 cells: Pre-adipocyte cell line responsive to insulin/IGF-1 stimulation

  • GIST (Gastrointestinal Stromal Tumor) cells: For studying IGF1R activation in cancer contexts

Stimulation protocols:

  • For acute activation: Treat serum-starved cells with 100 nM insulin or IGF-1 for 5-10 minutes at 37°C

  • For dose-response studies: Use IGF-1 concentration range of 1-100 nM with fixed time point

  • For time-course experiments: Treat with fixed concentration (typically 50-100 nM) for 2, 5, 10, 30, and 60 minutes

  • For inhibitor studies: Pre-treat with tyrosine kinase inhibitors before ligand stimulation

Considerations for specific research questions:

  • For cancer research: Compare phosphorylation levels between normal and cancer cell lines (phospho-IGF1R expression was detected in only 9% of cases without KIT mutation in GIST studies)

  • For cross-talk studies: Co-stimulate with insulin, IGF-1, and other growth factors

  • For mutational analysis: Compare wild-type cells with those expressing IGF1R variants (studies have used mutagenesis with clusters of alanine substitutions to identify surfaces important for receptor activation)

Research by Martins et al. demonstrated that analyzing phospho-IGF1R alongside MMP3 expression can provide insights into treatment response in gastrointestinal stromal tumors, with patients expressing either marker showing a 36% response rate to imatinib compared to 71% in those without expression .

How does phosphorylation at Tyr1165/Tyr1166 correlate with IGF1R activity and downstream signaling events?

The phosphorylation status of IGF1R at Tyr1165/Tyr1166 serves as a molecular barometer for receptor activation and correlates with downstream signaling through multiple pathways:

Correlation with kinase activity:

  • Phosphorylation at Tyr1165/Tyr1166 is essential but not sufficient for full kinase activation

  • Studies indicate that while every single phosphorylation increases kinase activity, all three tyrosine residues in the kinase activation loop have to be phosphorylated for optimal activity

  • Sequential phosphorylation occurs, with Tyr1165 typically phosphorylated first, followed by additional tyrosine residues

Downstream signaling pathways activated:

  • RAS/RAF/MAPK pathway: Leading to activation of ERK, p38, and JNK, promoting cell proliferation

  • PI3K/AKT pathway: Enhancing cell survival and metabolic regulation

  • These pathways ultimately lead to transcription of genes driving proliferation

Quantitative relationships:

  • The intensity of Tyr1165/Tyr1166 phosphorylation often shows dose-dependent relationships with ligand concentration

  • In cell-based studies, the ratio of phospho-IGF1R to total IGF1R provides a normalized measure of activation status

  • Time-course experiments typically show rapid phosphorylation (within minutes) followed by gradual dephosphorylation

Biological contexts:

  • In cancer research, phospho-IGF1R status may correlate with treatment response and prognosis (studies showed that phospho-IGF1R expression correlated with poorer response to imatinib in GIST patients)

  • In metabolic research, insulin resistance may be associated with altered patterns of IGF1R/IR phosphorylation

Understanding these correlations allows researchers to use phospho-IGF1R (Tyr1165/Tyr1166) detection as a reliable marker for receptor activation in various experimental and pathological contexts.

What are the methodological differences between studying IGF1R phosphorylation in cultured cells versus tissue samples?

Investigating IGF1R phosphorylation status presents distinct methodological challenges depending on whether the research involves cultured cells or tissue samples:

Cultured Cell Systems:

Advantages:

  • Controlled experimental conditions for precise stimulation timing and dose

  • Homogeneous cell populations reduce signal variability

  • Easy manipulation of receptor expression through transfection/transduction

  • Live-cell imaging possibilities with fluorescent phospho-antibodies

Methodological approaches:

  • Direct stimulation with defined concentrations of IGF-1 or insulin (typically 100 nM for 5-10 minutes)

  • Western blot analysis of whole cell lysates

  • Cell-based ELISA for higher throughput quantification

  • Immunofluorescence for subcellular localization of activated receptors

  • Flow cytometry for single-cell analysis of receptor activation

Tissue Sample Analysis:

Challenges:

  • Phosphorylation states may degrade rapidly post-collection

  • Heterogeneous cell populations complicate interpretation

  • Variable receptor expression across different cell types within the tissue

  • Limited control over pre-collection conditions

Methodological approaches:

  • Rapid tissue processing with phosphatase inhibitors is crucial

  • Tissue microarray construction for higher throughput analysis

  • Immunohistochemistry with phospho-specific antibodies

  • Scoring systems: In clinical studies, cases were scored as positive when at least 1% of cells showed cytoplasmic expression

  • Parallel analysis of downstream signaling markers

Comparative considerations:

In a GIST study (search result ), researchers constructed tissue microarrays from paraffin-embedded samples, applied heat-induced epitope retrieval with high-pH buffer, and used phospho-specific antibodies at 1:50 dilution, demonstrating that methodological optimization is essential for successful tissue analysis of phospho-IGF1R.

In what ways can phospho-IGF1R (Tyr1165/Tyr1166) antibodies be used to evaluate therapeutic interventions targeting the IGF1R pathway?

Phospho-IGF1R (Tyr1165/Tyr1166) antibodies serve as valuable tools for evaluating the efficacy of therapeutic interventions targeting the IGF1R pathway through multiple experimental approaches:

Assessing direct inhibitor effects:

  • Concentration-dependent inhibition curves: Measuring decreases in Tyr1165/Tyr1166 phosphorylation across inhibitor concentration ranges

  • Time-course analysis: Determining how rapidly inhibitors affect receptor activation status

  • Recovery kinetics: Monitoring re-activation of the receptor after inhibitor withdrawal

  • Resistance mechanisms: Identifying persistent phosphorylation despite inhibitor presence

Therapeutic monitoring applications:

  • Western blot analysis of patient-derived xenografts before and after treatment

  • Immunohistochemistry of tumor biopsies at baseline and on-treatment

  • Pharmacodynamic biomarker development using quantitative ELISA methods

Combined targeting strategies:

  • Evaluating synergistic effects between IGF1R inhibitors and other targeted agents

  • Assessing pathway reactivation mechanisms through phospho-proteomic approaches

  • Measuring compensatory activation of insulin receptor or other RTKs

Clinical correlations:

  • In GIST studies, phospho-IGF1R expression was associated with treatment response. Patients with phospho-IGF1R expression showed a 36% response rate to imatinib compared to 71% in those without expression

  • Progression-free survival analysis revealed that phospho-IGF1R expression may be associated with treatment outcomes, though multivariate analysis indicated other factors (disease-free interval, performance status, disease extension) were stronger prognostic indicators

Methodological approaches:

  • Pre-clinical validation using cell lines with varying IGF1R expression levels

  • Development of standardized protocols for phospho-IGF1R quantification in clinical samples

  • Multi-parameter analysis incorporating additional phosphorylation sites and downstream effectors

By systematically applying these approaches, researchers can comprehensively evaluate how therapeutic interventions impact IGF1R signaling and correlate biochemical responses with clinical outcomes.

How can phospho-IGF1R (Tyr1165/Tyr1166) detection be integrated with analysis of other receptor tyrosine kinases in cancer research?

Integrating phospho-IGF1R (Tyr1165/Tyr1166) detection with analysis of other receptor tyrosine kinases (RTKs) creates a comprehensive view of signaling networks in cancer research:

Multiplexed detection approaches:

  • Multi-color immunofluorescence: Simultaneous visualization of phospho-IGF1R alongside other activated RTKs (EGFR, HER2, etc.)

  • Reverse phase protein arrays (RPPA): Quantitative profiling of multiple phosphorylated receptors from limited sample material

  • Mass spectrometry-based phosphoproteomics: Unbiased detection of phosphorylation networks

  • Sequential immunoblotting: Stripping and reprobing membranes with different phospho-specific antibodies

Cross-talk analysis:

  • Co-immunoprecipitation studies to detect physical interactions between IGF1R and other RTKs

  • Inhibitor combination experiments to identify compensatory activation mechanisms

  • siRNA/CRISPR approaches to analyze signaling dependencies

Clinical research applications:

  • Tissue microarray analysis incorporating multiple phospho-RTK antibodies

  • Pattern recognition in RTK activation profiles across tumor subtypes

  • Correlation with genomic alterations (mutations, amplifications, deletions)

Case study from search results:
In GIST research by Martins et al., phospho-IGF1R (Y1316) analysis was integrated with KIT and PDGFRA mutation status and MMP3 expression :

  • Phospho-IGF1R was expressed in only 9% of cases without KIT mutation

  • MMP3 expression varied by genotype: 40% of PDGFRA-mutated, 6% of wild-type, and 10% of KIT-mutated patients

  • Combined analysis of both markers created an "immunophenotype" signature that correlated with treatment response

Data integration framework:

  • Hierarchical clustering of phospho-RTK patterns

  • Network analysis identifying central nodes and feedback mechanisms

  • Machine learning approaches to predict drug sensitivity based on phosphorylation profiles

This integrated approach enables researchers to understand IGF1R activation in the context of broader signaling networks, potentially revealing therapeutic vulnerabilities and resistance mechanisms in cancer.

What are the technical challenges in detecting transient phosphorylation events at Tyr1165/Tyr1166 following receptor activation?

Detecting transient phosphorylation events at IGF1R Tyr1165/Tyr1166 presents several technical challenges that researchers must address through careful experimental design:

Temporal dynamics challenges:

  • Rapid phosphorylation kinetics: IGF1R phosphorylation can occur within seconds to minutes after ligand binding

  • Transient nature: Dephosphorylation by phosphatases quickly counterbalances kinase activity

  • Asynchronous cellular responses: Not all cells in a population respond simultaneously

Methodological solutions:

  • Rapid sample processing: Immediate lysis in phosphatase inhibitor-containing buffers

  • Time-course experiments: Precise timing intervals (e.g., 0, 1, 2, 5, 10, 30 minutes)

  • Temperature control: Performing stimulations at lower temperatures (e.g., 16°C) to slow down enzyme kinetics

  • Cross-linking approaches: Chemical fixation immediately after stimulation

Detection sensitivity issues:

  • Low abundance of phosphorylated receptor relative to total receptor pool

  • Signal-to-noise challenges in heterogeneous samples

  • Antibody affinity and specificity limitations

Enhanced detection strategies:

  • Enrichment through immunoprecipitation before Western blot analysis

  • Proximity ligation assays for single-molecule sensitivity

  • Phospho-flow cytometry for single-cell resolution

  • Super-resolution microscopy for subcellular localization of activation events

Validation approaches:

  • Phosphatase treatment controls to confirm signal specificity

  • Peptide competition assays as demonstrated in search result

  • Correlation with downstream signaling events (e.g., AKT, ERK phosphorylation)

  • Genetic controls using kinase-dead receptor mutants

Quantification considerations:

  • Normalization to total receptor levels to account for expression differences

  • Internal loading controls specific for phospho-protein studies

  • Digital image analysis with appropriate dynamic range settings

  • Statistical approaches for time-series data

By addressing these challenges through rigorous methodology, researchers can accurately capture the dynamic nature of IGF1R phosphorylation and its implications for downstream signaling.

How do recent technological advances improve the detection and quantification of phospho-IGF1R (Tyr1165/Tyr1166) in research settings?

Recent technological advances have significantly enhanced our ability to detect and quantify phospho-IGF1R (Tyr1165/Tyr1166), providing researchers with more sensitive, specific, and informative methodologies:

Advanced antibody technologies:

  • Single-domain antibodies (nanobodies): Smaller size enables better epitope access

  • Recombinant antibody fragments: Consistent performance across batches

  • Monoclonal rabbit antibodies: Combining high specificity with superior affinity

  • Phospho-specific antibodies validated through comprehensive specificity testing (phosphopeptide competition, phosphatase treatment)

High-sensitivity detection systems:

  • Automated Western blot platforms: Increased reproducibility and quantitative capacity

  • Chemiluminescence substrates with femtogram sensitivity

  • Near-infrared fluorescent secondary antibodies: Enhanced linear dynamic range

  • Cell-based ELISA kits specifically designed for phospho-IGF1R detection

Single-cell analysis methods:

  • Mass cytometry (CyTOF): Simultaneous detection of multiple phospho-proteins

  • Imaging flow cytometry: Combining visual confirmation with quantitative analysis

  • Single-cell Western blot techniques: Protein analysis at individual cell level

  • Live-cell biosensors: Real-time monitoring of phosphorylation dynamics

Quantitative image analysis:

  • Automated high-content imaging: Subcellular localization and phosphorylation quantification

  • Machine learning algorithms: Unbiased pattern recognition in complex datasets

  • 3D tissue imaging: Spatial context of receptor activation in intact specimens

Multi-omics integration:

  • Correlation of phospho-proteomics with transcriptomics and metabolomics data

  • Systems biology approaches to model signaling networks

  • Computational methods to predict phosphorylation dynamics

Practical applications:

  • Phospho-IGF1R Cell-Based ELISA Kits allow for qualitative determination of IGF1R phosphorylation and multiple normalization methods

  • RayBio® Human Phospho-IGF1R (Y1165/1166) Kits provide semi-quantitative measurement in lysate samples

  • Tissue microarray approaches enable high-throughput analysis across multiple patient samples

These technological advances collectively improve the precision, throughput, and information content of phospho-IGF1R analysis, enabling researchers to address increasingly sophisticated questions about receptor biology and signaling dynamics.

What are the key differences between commercially available phospho-IGF1R (Tyr1165/Tyr1166) antibodies?

When selecting among commercially available phospho-IGF1R (Tyr1165/Tyr1166) antibodies, researchers should consider several distinguishing characteristics that impact experimental outcomes:

Immunogen design and specificity:

Antibody SourceImmunogen SequenceCross-ReactivitySpecificity Validation
Thermo Fisher (PA5-118536)Peptide sequence around phosphorylation site of tyrosine 1165/1166 (T-D-Y(p)-Y(p)-R-K)Human, Mouse, RatDetects endogenous levels when phosphorylated at specific sites
GeneTex (GTX25680)Not specified in search resultsHuman, Mouse, RatValidated through peptide competition assays
Sigma-Aldrich (SAB4300069)(T-D-YP-YP-R-K)Human, Mouse, RatWestern blot analysis of 293 cells
Millipore/Merck (ABE332)Not specifiedHuman, Pig, Mouse, Xenopus, Chicken, Zebrafish, RatCell-based assay and Western blot

Application validation:

AntibodyWestern BlotICC/IFIHCELISAFlow Cytometry
Fisher (PA537602)---
St John's (STJ90299)---
GeneTex (GTX25680)--
RayBiotech Kit----

Dilution recommendations:

  • Western blot: Ranges from 1:500-1:2000 depending on manufacturer

  • Immunocytochemistry: Typically 1:100-1:200

  • ELISA: Generally more dilute, up to 1:20000 for some antibodies

Storage conditions and formulations:

  • Most antibodies are supplied in PBS with 50% glycerol and preservatives (0.02% sodium azide)

  • Storage temperature recommendations consistently indicate -20°C

  • Concentration typically 1 mg/mL

Additional considerations:

  • Some antibodies recognize both IR and IGF1R phosphorylation due to sequence homology

  • Others are specifically designed to recognize only IGF1R phosphorylation

  • Antibodies may differ in their recognition of dual (Tyr1165/1166) versus triple phosphorylation sites

These differences highlight the importance of selecting antibodies based on specific experimental requirements and validating their performance in your particular system before conducting extensive studies.

How can researchers distinguish between IGF1R and insulin receptor phosphorylation in experimental systems?

Distinguishing between IGF1R and insulin receptor (IR) phosphorylation presents a significant challenge due to their structural homology, but several strategies can help researchers achieve specificity:

Antibody-based discrimination strategies:

  • Specific phospho-epitope antibodies:

    • Select antibodies validated against specific phosphorylation sites that differ between IGF1R and IR

    • Some antibodies are developed to recognize both receptors (e.g., those recognizing IR/IGF1R at Tyr1162/Tyr1163 of IR and Tyr1135/Tyr1136 of IGF1R)

    • Others target IGF1R-specific phosphorylation sites or conformations

  • Sequential immunoprecipitation:

    • First immunoprecipitate with receptor-specific antibodies (targeting non-conserved regions)

    • Then probe with phospho-specific antibodies to analyze activation state

  • Isoform-specific detection:

    • Combine with antibodies against unique regions (e.g., C-terminal domains)

    • Use antibodies that recognize receptor-specific phosphorylation patterns

Experimental design approaches:

  • Selective stimulation:

    • Use IGF-1 at concentrations that preferentially activate IGF1R over IR

    • Apply insulin analogs with reduced IGF1R binding capacity

  • Genetic manipulation:

    • siRNA knockdown or CRISPR knockout of one receptor

    • Overexpression of one receptor in cells with low endogenous expression

    • Expression of epitope-tagged receptors for specific isolation

  • Specific inhibition:

    • Pre-treatment with receptor-selective inhibitors

    • Blocking antibodies that specifically prevent activation of one receptor

Validation methodologies:

  • Cross-validation:

    • Compare results using multiple antibodies targeting different epitopes

    • Verify with receptor-specific functional assays

  • Control experiments:

    • Use cell lines expressing only IGF1R or IR

    • Include phosphatase-treated samples as negative controls

    • Peptide competition assays with receptor-specific phosphopeptides

Example from search results: GeneTex antibody GTX25680 was validated through peptide competition analysis, showing that only the immunogen phosphopeptide blocks the signal in Western blot of insulin-stimulated CHO-T cells, demonstrating its specificity for phosphorylated receptor .

By combining these approaches, researchers can achieve reliable discrimination between IGF1R and insulin receptor phosphorylation, essential for accurately characterizing their distinct and overlapping signaling functions.

What are the critical controls needed to ensure valid interpretation of phospho-IGF1R (Tyr1165/Tyr1166) data?

Robust experimental design for phospho-IGF1R (Tyr1165/Tyr1166) detection requires comprehensive controls to ensure valid data interpretation:

Positive and negative sample controls:

Control TypePurposeImplementation
Positive controlVerify antibody functionalityInsulin/IGF-1 stimulated cells (e.g., 293 cells, MCF-7 cells treated with 100 nM for 5-10 minutes)
Negative controlEstablish baseline/backgroundSerum-starved unstimulated cells
Phosphatase-treatedConfirm phospho-specificitySample aliquot treated with lambda phosphatase
Receptor knockout/knockdownValidate signal specificityCRISPR-Cas9 or siRNA-treated cells

Antibody validation controls:

  • Peptide competition assays:

    • Pre-incubate antibody with:

      • Phosphorylated peptide immunogen (should block signal)

      • Non-phosphorylated version of same peptide (should not block)

      • Generic phosphotyrosine-containing peptide (should not block)

    • As demonstrated in GeneTex antibody validation, only the specific phosphopeptide immunogen should block the signal

  • Antibody dilution series:

    • Determine optimal concentration for signal-to-noise ratio

    • Typically 1:500-1:2000 for Western blot applications

  • Secondary antibody-only controls:

    • Identify non-specific background binding

Experimental treatment controls:

  • Dose-response controls:

    • Titration of IGF-1/insulin (typically 1-100 nM)

    • Establish relationship between stimulus intensity and phosphorylation

  • Time-course controls:

    • Multiple time points after stimulation (e.g., 0, 2, 5, 10, 30, 60 minutes)

    • Capture transient phosphorylation dynamics

  • Kinase inhibitor controls:

    • IGF1R-specific inhibitors to confirm signal origin

    • Broad-spectrum tyrosine kinase inhibitors as positive controls for inhibition

Normalization controls:

  • Total protein controls:

    • Total IGF1R antibody on parallel samples/stripped blots

    • Essential for distinguishing between increased phosphorylation versus increased receptor expression

  • Loading controls:

    • For Western blot: Beta-actin, GAPDH, or total protein stains

    • For cell-based assays: Crystal violet staining for cell number normalization

  • Multiple normalization approaches:

    • As described in search result , cell-based ELISAs can employ anti-GAPDH antibody as internal control, Crystal Violet whole-cell staining for cell density, and anti-total IGF1R antibody for expression normalization

Implementing these controls ensures that changes in phospho-IGF1R signal genuinely reflect biological responses rather than technical artifacts, and provides the framework for accurate quantification and interpretation of results.

How does IGF1R phosphorylation status correlate with cancer progression and treatment response?

The phosphorylation status of IGF1R at Tyr1165/Tyr1166 has emerged as a potentially significant biomarker in cancer research, with implications for disease progression and therapeutic response:

Correlations with cancer progression:

Studies have shown variable associations between phospho-IGF1R status and cancer progression markers:

  • In gastrointestinal stromal tumors (GISTs), phospho-IGF1R expression patterns vary by genetic background:

    • Expressed in only 9% of cases without KIT mutation

    • Frequently associated with specific genetic alterations

  • Activation of the IGF1R pathway has been implicated in:

    • Promoting cancer cell proliferation and survival

    • Enhancing metastatic potential

    • Contributing to tumor microenvironment modulation

    • Mediating resistance to various targeted therapies

Treatment response correlations:

In the GEIS-19 study examining GIST patients:

  • Response rate was observed in only 36% (4/11) of patients with phospho-IGF1R or MMP3 expression

  • Comparatively, 71% (56/78) of patients without expression responded to treatment

  • This difference was statistically significant (p = 0.025)

Progression-free survival analysis:

MarkerMedian PFS95% CIp-value
Phospho-IGF1R+ or MMP3+14.1 months0-29.80.33 (non-significant)
Phospho-IGF1R- and MMP3-37.1 months25.3-48.9

While the trend suggested poorer outcomes with phospho-IGF1R expression, this did not reach statistical significance in multivariate analysis .

Multivariate analysis findings:

Disease-free interval, performance status, extension of disease, and genotype emerged as stronger prognostic factors than phospho-IGF1R expression in the multivariate analysis .

These findings suggest that while phospho-IGF1R status correlates with treatment response in some contexts, its prognostic significance may depend on tumor type, genetic background, and therapeutic approach. The results highlight the importance of integrating phospho-IGF1R analysis with other molecular and clinical parameters for comprehensive patient stratification.

What methodological considerations are important when using phospho-IGF1R (Tyr1165/Tyr1166) antibodies in biomarker development?

Developing phospho-IGF1R (Tyr1165/Tyr1166) as a clinically relevant biomarker requires addressing several methodological considerations to ensure reliability, reproducibility, and clinical utility:

Tissue handling and pre-analytical variables:

  • Rapid tissue fixation is critical: Phosphorylation states degrade quickly (minutes)

  • Standardized fixation protocols: Typically 10% neutral buffered formalin for 24-48 hours

  • Controlled cold ischemia time: Ideally <30 minutes from excision to fixation

  • Consistent processing methods: Embedding, sectioning thickness (3-5 μm)

  • Application of phosphatase inhibitors during tissue handling when possible

Antibody validation for clinical applications:

  • More rigorous validation than research-only applications

  • Multi-platform concordance testing (IHC, Western blot, ELISA)

  • Analytical specificity determination: Cross-reactivity profiling

  • Reproducibility assessment: Inter-laboratory and inter-observer studies

  • Lot-to-lot consistency monitoring for long-term studies

Immunohistochemistry optimization:

From the GIST study methodology :

  • Epitope retrieval: Heat-induced with high-pH buffer for phospho-IGF1R antibodies

  • Antibody dilution: 1:50 dilution following optimization

  • Detection system: DAB Map detection kit

  • Counterstaining: Hematoxylin for nuclear visualization

  • Scoring system: Cases considered positive when ≥1% of cells showed expression

Quantification and scoring approaches:

  • Qualitative scoring: Positive/negative based on defined threshold (≥1% in GIST study)

  • Semi-quantitative methods: H-score, Allred score, or similar systems

  • Digital pathology: Automated quantification of staining intensity and distribution

  • Multiplex IHC: Co-localization with total IGF1R and other markers

Clinical correlation considerations:

  • Prospective vs. retrospective study design

  • Sample size determination through power analysis

  • Selection of appropriate clinical endpoints (response, PFS, OS)

  • Integration with other established biomarkers

  • Standardized reporting formats (REMARK guidelines)

Challenges and solutions:

  • Heterogeneity: Assess multiple tumor regions when possible

  • Background staining: Optimize blocking and implement appropriate controls

  • Interpretation variability: Train multiple observers and assess concordance

  • Cut-point determination: Use statistical methods (ROC analysis) to establish clinically relevant thresholds

Addressing these methodological considerations is essential for translating phospho-IGF1R detection from a research tool to a clinically actionable biomarker with potential applications in patient stratification and therapeutic decision-making.

How might emerging technologies enhance our understanding of IGF1R phosphorylation dynamics?

Emerging technologies are poised to revolutionize our understanding of IGF1R phosphorylation dynamics by enabling higher resolution, real-time analysis across multiple scales:

Advanced imaging technologies:

  • Super-resolution microscopy: Techniques like STORM, PALM, and SIM can visualize individual phosphorylated receptors with nanometer-scale precision, potentially revealing spatial clustering patterns and co-localization with signaling partners

  • Live-cell FRET biosensors: Genetically encoded sensors that undergo conformational changes upon IGF1R phosphorylation, allowing real-time visualization of activation dynamics with subcellular resolution

  • Expansion microscopy: Physical enlargement of specimens to visualize receptor complexes and phosphorylation patterns below the diffraction limit

  • Light-sheet microscopy: For rapid 3D imaging of phospho-IGF1R distribution in organoids and tissue sections

Single-cell analysis platforms:

  • Single-cell phospho-proteomics: Mass spectrometry-based techniques capable of quantifying multiple phosphorylation sites in individual cells

  • Single-cell Western blotting: Miniaturized protein separations from individual cells to assess phospho-IGF1R heterogeneity

  • Mass cytometry (CyTOF): Simultaneous detection of multiple phosphorylated signaling proteins across thousands of single cells using metal-tagged antibodies

  • Spatial transcriptomics: Correlating phospho-protein states with gene expression patterns in intact tissue contexts

Computational and systems biology approaches:

  • Machine learning algorithms: Pattern recognition in complex phosphorylation datasets to identify novel regulatory relationships

  • Mathematical modeling: Differential equation-based models of receptor phosphorylation kinetics under various conditions

  • Network analysis: Integration of phospho-IGF1R data with broader signaling networks to understand system-level behaviors

  • Digital pathology integration: Automated quantification and spatial analysis of phospho-IGF1R in clinical specimens

Molecular manipulation technologies:

  • Optogenetics: Light-controlled activation of IGF1R to precisely manipulate phosphorylation with high spatiotemporal control

  • Chemical genetics: Engineered IGF1R variants that can be selectively activated by synthetic ligands

  • CRISPR-based screening: Systematic identification of genes affecting IGF1R phosphorylation dynamics

  • Proximity labeling: BioID or APEX2 fusions to phospho-IGF1R to catalog proteins associated with the activated receptor

By integrating these emerging technologies, researchers will gain unprecedented insights into the temporal dynamics, spatial organization, and molecular contexts of IGF1R phosphorylation events, potentially revealing new therapeutic opportunities and biomarker applications.

What novel therapeutic strategies might emerge from better understanding of IGF1R phosphorylation mechanisms?

Enhanced understanding of IGF1R phosphorylation mechanisms is driving the development of novel therapeutic strategies with potential applications across multiple disease contexts:

Targeted inhibition approaches:

Phosphatase activation strategies:

  • Phosphatase-activating compounds: Molecules that enhance the activity of phosphatases specifically acting on IGF1R

  • Phosphatase-targeting protein-drug conjugates: Directing phosphatases to IGF1R to promote dephosphorylation

  • Inhibitors of phosphatase inhibitors: Compounds that release phosphatases from natural inhibitory constraints

Differential pathway modulation:

  • Biased signaling inducers: Compounds that selectively activate beneficial pathways downstream of IGF1R while inhibiting detrimental ones

  • Temporal modulation: Pulsatile inhibition strategies based on phosphorylation dynamics

  • Synthetic receptors: Engineered IGF1R variants with modified phosphorylation sites for selective pathway activation

Combination therapy approaches:

  • Vertical pathway inhibition: Combining IGF1R phosphorylation inhibitors with downstream pathway blockers

  • Horizontal pathway targeting: Simultaneously inhibiting IGF1R and compensatory RTKs (e.g., insulin receptor)

  • Immunotherapy combinations: Exploiting potential synergies between immune checkpoint inhibitors and IGF1R phosphorylation inhibitors

Precision medicine applications:

  • Phosphorylation-based patient stratification: Selecting patients based on IGF1R phosphorylation status (GIST studies showed differential treatment response based on phospho-IGF1R expression)

  • Adaptive therapy: Modifying treatment based on changes in phosphorylation patterns during therapy

  • Predictive biomarkers: Using phospho-IGF1R alongside other markers to predict response to various therapies

Translational opportunities:

  • Reducing insulin resistance: Selective modulation of IGF1R/IR phosphorylation for metabolic disorders

  • Cancer therapy: Targeting aberrant IGF1R phosphorylation in malignancies with pathway activation

  • Neurodegenerative disorders: Modulating IGF1R phosphorylation for neuroprotection

  • Aging interventions: Calibrating IGF1R signaling to potentially extend healthspan

These emerging therapeutic strategies represent promising directions for translating basic research on IGF1R phosphorylation into clinical applications with potential impact across multiple disease domains.

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 2025 TheBiotek. All Rights Reserved.