Recombinant Human Insulin-like growth factor I (IGF1) (Active) (GMP)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

Receptor Binding:

  • Primary target: IGF1 receptor (IGF1R)

  • Secondary targets: Insulin receptor, integrins (ITGAV:ITGB3, ITGA6:ITGB4)

Key Signaling Pathways:

  1. PI3K-AKT Pathway: Promotes cell survival and glucose uptake

  2. Ras-MAPK Pathway: Stimulates proliferation and differentiation

  3. Cross-talk: Modulates steroid hormone activity and bone metabolism

Pharmacokinetics:

  • Half-life: ~15 hours (subcutaneous administration)

  • Plasma stability: Enhanced by IGF-binding proteins (IGFBPs)

Clinical and Research Applications

Therapeutic Uses:

  • Growth Disorders: Increases height velocity in IGF1-deficient patients (3.8 cm/yr → 6.6 cm/yr)

  • Metabolic Regulation: Improves insulin sensitivity in diabetes models

  • Tissue Repair: Enhances muscle regeneration post-injury

Oncology Research:

  • Stimulates proliferation in MCF-7 breast cancer cells (EC50: 2-14 ng/mL)

  • Upregulates anti-apoptotic proteins via AKT signaling

Neuroscience Applications:

  • Required for olfactory bulb function (Ca²⁺-dependent exocytosis)

  • Modulates synaptic maturation in CNS development

Key Research Findings

Clinical Trial Data (Source ):

  • Dose Response: 40 μg/kg vs. 80 μg/kg s.c.

    • Peak IGF1 levels: 341 μg/L vs. 794 μg/L

    • IGFBP-3 increase: 5.8 mg/L → 9 mg/L

    • GH suppression: 342 mU/L → 40 mU/L

In Vitro Performance:

  • Potency: Stimulates [1-14C]-2-deoxy-D-glucose transport at 0.2-20 ng/mL

  • Specific Activity: >5×10⁵ IU/mg in MCF-7 proliferation assays

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered concentrated solution in PBS, pH 7.0.
Form
Liquid or Lyophilized powder
Lead Time
Typically, we can ship the products within 1-3 working days of receiving your order. Delivery times may vary depending on the method of purchase or location. Please consult your local distributors for specific delivery times.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please communicate with us in advance as additional fees will apply.
Shelf Life
Shelf life is influenced by factors such as storage conditions, buffer components, temperature, and the protein's inherent stability.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The lyophilized form has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
IBP1; IGF I; IGF IA; IGF IB; IGF-I; Igf1; IGF1_HUMAN; IGF1A; IGFI; IGFIA; Insulin like growth factor 1 (somatomedin C) ; Insulin like growth factor 1; Insulin like growth factor IA; Insulin like growth factor IB; Insulin-like growth factor I; Mechano growth factor; MGF; OTTHUMP00000195080; OTTHUMP00000195081; OTTHUMP00000195082; OTTHUMP00000195083; OTTHUMP00000195084; Somatomedia C; Somatomedin C; Somatomedin-C
Datasheet & Coa
Please contact us to get it.
Expression Region
49-118aa
Mol. Weight
7.6 kDa
Protein Length
Full Length of Mature Protein
Purity
> 98 % by SDS-PAGE and HPLC analyses.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Insulin-like growth factors, isolated from plasma, exhibit structural and functional similarities to insulin but possess significantly higher growth-promoting activity. IGF1 may serve as a physiological regulator of [1-14C]-2-deoxy-D-glucose (2DG) transport and glycogen synthesis in osteoblasts. It stimulates glucose transport in bone-derived osteoblastic (PyMS) cells and is effective at significantly lower concentrations than insulin, not only in terms of glycogen and DNA synthesis but also in enhancing glucose uptake. IGF1 may play a role in synapse maturation. Ca(2+)-dependent exocytosis of IGF1 is essential for sensory perception of smell in the olfactory bulb. IGF1 acts as a ligand for IGF1R. It binds to the alpha subunit of IGF1R, leading to the activation of the intrinsic tyrosine kinase activity. This activity autophosphorylates tyrosine residues in the beta subunit, initiating a cascade of downstream signaling events, ultimately activating the PI3K-AKT/PKB and Ras-MAPK pathways. IGF1 binds to integrins ITGAV:ITGB3 and ITGA6:ITGB4. Its binding to integrins and subsequent ternary complex formation with integrins and IGFR1 are crucial for IGF1 signaling. IGF1 induces the phosphorylation and activation of IGFR1, MAPK3/ERK1, MAPK1/ERK2, and AKT1.
Gene References Into Functions
  1. SNX6 knockdown resulted in a dramatic reduction in IGF1-mediated ERK1/2 phosphorylation but did not affect IGF1R internalization. PMID: 29530981
  2. We observed that elevated production of insulin-like growth factor 1 by polyploid ASCs rendered them more potent in promoting tumor growth in vitro. PMID: 30185144
  3. No up-regulation of Wnt10A and IGF-1 mRNA was observed with 1,550-nm Er:Glass fractional laser treatment of androgenetic alopecia. PMID: 30096107
  4. Significant negative or positive correlations were observed between IGF-1 concentrations and impairments on several EDI-2 subscales (drive for thinness, body dissatisfaction, interoceptive awareness, sense of ineffectiveness, interpersonal distrust, maturity fear) and on SCL-90 subitems (depression, hostility, obsessivity compulsivity, anxiety), suggesting a possible hormonal modulatory effect on specific aspects of eating behaviors. PMID: 29179911
  5. This article provides a review of the tumor-specific molecular signatures of IGF-1-mediated epithelial-mesenchymal transition in breast, lung, and gastric cancers. [review] PMID: 30111747
  6. Results provide evidence for the role of IGF-I: matrix protein interactions in cell growth, migration, and melanoma progression through complex formation with IGF binding proteins. PMID: 29330502
  7. The molecular interactions of IGF1-IGF1R binding have been elucidated. PMID: 29483580
  8. Genetic association and nutrigenomic studies in a population of postmenopausal women in China: Data confirm that dietary acid load is associated with postmenopausal osteoporosis; an SNP in IGF1 (rs35767) is not associated with this relationship in the population studied. PMID: 30018240
  9. Low IGF-I levels are associated with non-alcoholic fatty liver disease. PMID: 29395967
  10. Low IGF1 expression is associated with retinopathy of prematurity. PMID: 29274846
  11. Low IGF-I levels are associated with dry eye syndrome. PMID: 29452886
  12. Serum IGF-1 is associated with greater muscle mass, higher bone mineral density, and improved handgrip performance in both genders among community-dwelling middle-aged and older adults in Taiwan. PMID: 28351218
  13. Insulin-like growth factor 1 (IGF1) is a direct target of miR-1827. PMID: 28387248
  14. Results indicate that the expression of IGF-1 is up-regulated in cumulus cells (CCs) of women with polycystic ovary syndrome (PCOS), which was inversely proportional to the expression level of miR-323. Furthermore, IGF1 3'UTR is targeted by miR-323. PMID: 30300681
  15. A study in hepatocellular carcinoma cell line elucidated a new mechanism in which IGF-1 induced epithelial-mesenchymal transition through the regulation of survivin and a downstream pathway. PMID: 29989646
  16. This study assessed the influence of IGF-1 and BMP-7 at different concentrations on the osteogenic differentiation of two human MSC-subtypes, isolated from reaming debris (RMSC) and iliac crest bone marrow (BMSC). PMID: 29874864
  17. AMH, IGF1, and leptin levels in follicular fluid have no relation to the fertility disorders caused by endometriosis or fallopian tube damage, although they are biomarkers for anovulatory fertility disorders. PMID: 29595066
  18. Pituitary growth hormone secretory capacity is not related to IGF-1 levels. PMID: 29098662
  19. Peripheral totalIGF-1 and IGFBP-3 were associated with better performance in attention, visuospatial, and global cognitive domains, independent of gait speed. PMID: 29547749
  20. The CA repeat polymorphism of the P1 promoter of the IGF1 gene was associated with strength predispositions in the homozygous and non-carriers groups. In the heterozygous group, it was speed-strength aptitudes. PMID: 29543920
  21. IGF1 levels in systemic lupus erythematosus patients decline with age at a similar rate as in healthy controls, are associated with positive metabolic effects, and despite a moderating effect on B, NK, and CD8+ cells, do not affect clinical disease activity or severity. PMID: 29385899
  22. Results indicate that IGF-1 gene polymorphisms play crucial roles in the histopathological progression of IgA Nephropathy in the Chinese Han population. PMID: 29402846
  23. Low IGF1 level is associated with steatosis in Pituitary Diseases. PMID: 29065431
  24. They retained the main IGF-1R-related properties, but the hormones with His49 in IGF-1 and His48 in IGF-2 showed significantly higher affinities for IR-A and for IR-B, being the strongest IGF-1- and IGF-2-like binders of these receptors ever reported. PMID: 29608283
  25. The mechanism of activation of SGK3 by IGF1 via the class 1 and class 3 phosphatidylinositol 3-kinases has been described. PMID: 29150437
  26. IGF1 is associated with giant cell tumor of bone recurrence, which may serve as a biomarker for giant cell tumor of bone recurrence. PMID: 29651441
  27. miR-19b is decreased in PCOS granulosa cells, and miR-19b could be a granulosa cell proliferation inhibitor. miR-19b-mediated cell proliferation may act via directly targeting IGF-1. PMID: 29363717
  28. A report describes the deregulation of IGF1/IGFBP3 expression in breast cancer, correlating with neoplasm staging and histologic grade. PMID: 30084561
  29. A study observed that the IGF1 level was higher in human epithelial ovarian cancer (EOC) specimens than in benign ovarian tumor specimens. Further analysis showed that a higher level of IGF1 was related to more advanced clinical stage and liver metastasis. Additionally, up-regulation of IGF1 by tumor-associated macrophages was shown to promote the proliferation and migration of EOC cell lines. PMID: 29251331
  30. Overexpression of circHIPK3 increased the expression levels of IGF1, and knock-down reduced it. PMID: 28738961
  31. Women who remained obese had an increased cGP/IGF-1 ratio (p = 0.006) only. An increase in the cGP/IGF-1 ratio is associated with obesity, but not hypertension. Changes in IGFBP-3 and/or the cGP/IGF-1 ratio are associated with weight changes. The data suggest a role for cGP in obesity through autocrine regulation of IGF-1. PMID: 29921371
  32. IGF-1 exerts antioxidant, anti-inflammatory, and protective effects on the central nervous system. (Review) PMID: 29975480
  33. Five single nucleotide polymorphisms (SNPs) of IGF-1 (rs6214, rs6218, rs35767, rs5742612, and rs5742714) were genotyped. DNA was extracted from peripheral blood and analyzed for SNP genotyping using PCR. rs6218 had a predictive role for the susceptibility and progression of osteosarcoma. The TC and CC genotypes of rs6218 indicated a higher risk of osteosarcoma; associated with later stage and elevated risk of osteosarcoma. PMID: 29232358
  34. Bioinformatics analysis of 982 lung cancer patients revealed that higher expression of TNF- alpha was associated with a low risk of cancer progression, while overexpression of IGF-1 was correlated with a high risk. Collectively, these results reveal that the cytokines in the tumor microenvironment differentially modulate radiation therapy through various signaling mechanisms. PMID: 27344406
  35. Data suggest that lower IGF1 serum levels in aging are associated with lower handgrip strength and worse physical performance, but less recurrent falls, especially in men. This longitudinal study was conducted in the Netherlands. PMID: 29789408
  36. Data suggest that the R1353H point mutation constitutes an activating IGF1R variant that can result in extreme tall height with very low levels of serum IGFI; the mutation was observed in the proband, his mother, and his 3 sons. However, only the proband (43 y/o) and eldest son (18 y/o) exhibit extreme tall height. [CASE REPORT] PMID: 29789409
  37. IGF-1 treatment activated protein kinase B (AKT), which may inhibit autophagy via the AKT/mammalian target of rapamycin signaling pathway. Following inhibition of autophagy, drug-resistant cells became sensitive to apoptosis induced by 5-fluorouracil. PMID: 29257307
  38. Consistent with its enhanced expression in Laron syndrome, we provide evidence that TXNIP gene expression is negatively regulated by IGF1. PMID: 29339473
  39. In elderly women with low-energy distal radius fractures, an association between IGF1 and the lowest measures of bone mineral density was found, indicating that low IGF1 could be an indirect risk factor for fractures. PMID: 29178313
  40. Expression of miR-30a-3p was significantly increased in the placentas of patients with preeclampsia. miR-30a-3p might be involved in the pathogenesis of preeclampsia by targeting IGF-1 and regulating the invasion and apoptosis of trophoblast cells. PMID: 29155142
  41. Results suggest that the different isoforms of the PI3K p110 subunit could be therapeutic targets for primary and metastatic colon cancer and that regulation of the NRD1/ADAM signaling pathway controls lipogenesis-mediated EMT in IGF-1-stimulated colon cancer cells. PMID: 28819788
  42. Demonstrate the uptake of fluorescently labeled IGF-I into skeletal growth plates of live mice using multiphoton microscopy. PMID: 28798204
  43. This study provides evidence that the IGF-1/IGF-1R/hsa-let-7c axis can control the odonto/osteogenic differentiation of IGF-1-treated stem cells from apical papilla via the regulation of JNK and p38 MAPK signaling pathways. PMID: 27833148
  44. Circulating sex steroids, prolactin, insulin-like growth factor (IGF) I, IGF-binding protein 3, and sex hormone-binding globulin (SHBG) were evaluated using backward elimination separately in women pre- and postmenopausal at blood collection. PMID: 28246273
  45. An association between serum IGFBP-1 and IGF-I levels with advanced fibrosis in non-alcoholic fatty liver disease patients was found. PMID: 28927302
  46. Findings suggest that the IGF1 polymorphism rs5742714 may be a genetic predictor of susceptibility and prognosis of renal cell carcinoma. PMID: 27976731
  47. The results of the study may help to inform future health interventions that utilize physical activity as a means to improve cognitive development in children, adolescents, and adults. Additionally, the study may assist in determining whether the putative effects occur via modification of plasma IGF-1 or BDNF concentrations. PMID: 29094050
  48. Data show the differential expression of insulin-like growth factor 1 (IGF-I) transcripts isoforms in bladder cancer, revealing a distinct suppression of IGF-IEc. PMID: 29848696
  49. Peripheral blood aspirates overexpressing IGF-I via rAAV gene transfer undergo enhanced chondrogenic differentiation processes. PMID: 28467017
  50. Human IGF-I propeptides and mutants were overexpressed in bovine articular chondrocytes. Secreted IGF-I propeptides stimulated articular chondrocyte biosynthetic activity as much as mature IGF-I. Of the 3 IGF-I propeptides, only proIGF-IA strongly bound to heparin, depending on N-glycosylation at Asn92 in the EA peptide. PMID: 29174671

Show More

Hide All

Database Links

HGNC: 5464

OMIM: 147440

KEGG: hsa:3479

STRING: 9606.ENSP00000302665

UniGene: Hs.160562

Involvement In Disease
Insulin-like growth factor I deficiency (IGF1 deficiency)
Protein Families
Insulin family
Subcellular Location
Secreted.

Q&A

What is the molecular structure of recombinant human IGF-I and how does it compare to native IGF-I?

Recombinant human IGF-I is a globular protein containing 70 amino acids with a calculated molecular weight of 7.6 kDa. It contains three intramolecular disulfide bonds that are critical for maintaining its three-dimensional structure and biological activity . The amino acid sequence and structural properties of recombinant human IGF-I are identical to those of endogenous human IGF-I, making it suitable for experimental research applications. Mature human IGF-I also demonstrates 100% homology with bovine and porcine proteins, which has important implications for comparative studies across species .

When designing experiments with recombinant IGF-I, researchers should consider that proper storage and handling conditions are essential to maintain the integrity of disulfide bonds and prevent protein aggregation. Most commercially available preparations require reconstitution according to manufacturer specifications to ensure optimal biological activity.

How does IGF-I differ functionally from insulin and growth hormone?

While IGF-I shares structural similarities with insulin, it demonstrates significantly higher growth-promoting activity. A key functional difference between rhIGF-I and recombinant human growth hormone (rhGH) is that rhIGF-I exhibits hypoglycemic activity similar to insulin, whereas rhGH opposes insulin action and is diabetogenic . This distinction has important implications for experimental design and potential therapeutic applications.

  • IGF-I has a longer half-life than GH, making it less susceptible to fluctuations and a more stable biomarker

  • IGF-I circulates bound to specific binding proteins (IGFBPs) that modulate its bioavailability and activity

  • IGF-I can stimulate glucose transport at much lower concentrations than insulin, particularly in bone-derived osteoblastic cells

What are the primary cellular responses induced by IGF-I?

IGF-I induces a diverse array of cellular responses including:

  • Cell proliferation

  • Cell differentiation

  • Cell migration

  • Cell survival and anti-apoptotic effects

These effects are tissue-dependent and context-specific. For example, IGF-I stimulates the proliferation of various cell types in muscle, bone, and cartilage tissue while enhancing glucose uptake in metabolically active cells . IGF-I is also recognized as one of the most potent activators of the AKT signaling pathway, which promotes proliferation and inhibits programmed cell death .

When designing experiments to evaluate these responses, researchers should consider using multiple complementary assays to capture the full spectrum of IGF-I bioactivities, as single readouts may not reflect the complexity of IGF-I's effects on cellular physiology.

What is the molecular mechanism of IGF-I receptor activation?

The activation of the type I insulin-like growth factor receptor (IGF1R) follows a precise molecular sequence that begins with ligand binding. Current research indicates that in the absence of ligand, the IGF1R maintains an autoinhibited state characterized by separation of the transmembrane (TM) domains . This configuration prevents spontaneous receptor activation.

The molecular activation process occurs as follows:

  • IGF-I binds specifically to the α subunit of IGF1R on the cell surface

  • Ligand binding induces a conformational change in the β subunit

  • This conformational change releases constraints that normally keep the TMs separated

  • The TMs subsequently associate, enabling the intrinsic propensity of the kinase regions to autophosphorylate

  • Autophosphorylation of the receptor triggers activation of receptor tyrosine kinase activity

Molecular dynamics simulations have revealed that IGF1R TMs can form stable dimers where the two TM helices cross each other opposite a conserved proline residue (P911) . This structural insight provides important mechanistic understanding of how ligand binding translates to receptor activation across the membrane.

What are the critical downstream signaling pathways activated by IGF-I and how are they experimentally distinguished?

IGF-I receptor activation initiates several distinct but interconnected signaling cascades. The activated receptor phosphorylates multiple substrates, primarily insulin receptor substrates (IRSs) and Src homology collagen (SHC) . These phosphorylated substrates serve as docking sites for SH2 domain-containing signaling molecules, including:

  • The 85 kDa regulatory subunit (p85) of phosphatidylinositol 3-kinase (PI 3-kinase)

  • Growth factor receptor-bound 2 (GRB2)

  • SH2-containing protein tyrosine phosphatase 2 (SHP2/Syp)

These interactions lead to the activation of two primary downstream signaling pathways:

PathwayKey ComponentsPrimary Cellular Responses
PI 3-kinase PathwayPI3K, AKT/PKB, mTORCell survival, metabolism, protein synthesis
Ras-MAPK PathwayRas, Raf, MEK, ERKCell proliferation, differentiation, gene expression

To experimentally distinguish between these pathways, researchers commonly employ specific inhibitors (e.g., wortmannin for PI3K, U0126 for MEK), genetic knockdown using siRNA techniques, or genetic knockout via conventional methods or CRISPR-Cas9 systems . These approaches allow for the assessment of necessity or sufficiency of each pathway for specific IGF-I bioactivities.

How do IGF binding proteins (IGFBPs) modulate IGF-I activity and how should this be accounted for in experimental designs?

In circulation, IGF-I predominantly binds to specific binding proteins (IGFBPs) which serve two critical functions: prolonging the half-life of IGF-I and facilitating its delivery to target tissues . This binding substantially impacts both the bioavailability and activity of IGF-I, creating an additional layer of regulation that must be considered in experimental designs.

The presence of IGFBPs creates significant challenges for IGF-I measurement and activity assessment:

  • IGFBPs can interfere with IGF-I immunoassays, potentially leading to inaccurate quantification

  • The IGFBP binding affects the free fraction of IGF-I available for receptor activation

  • Some IGFBPs have intrinsic biological activities independent of IGF-I

To account for these factors in experimental designs, researchers should consider:

  • Using techniques that separate IGF-I from its binding proteins prior to measurement

  • Implementing modern mass spectrometric methods that minimize interference from IGF-I variants

  • Including controls that assess both total and free IGF-I levels

  • When using recombinant IGF-I in cell culture systems, accounting for the presence of IGFBPs secreted by the cells themselves

These considerations are essential for accurate interpretation of IGF-I-related experimental data, particularly when translating between in vitro and in vivo systems.

What are the challenges in standardizing IGF-I measurement methods and how can researchers address them?

Despite its importance as a biomarker, IGF-I measurement faces significant standardization challenges that researchers must address to ensure reliable and comparable results. Key challenges include:

  • Interference from IGFBPs in immunoassays

  • Presence of IGF-I variants that may affect quantification

  • Variability in reference intervals across different assay platforms

  • Pre-analytical factors that influence IGF-I levels

To address these challenges, researchers should implement the following methodological approaches:

  • Employ techniques that efficiently separate IGF-I from IGFBPs before measurement

  • Consider using mass spectrometric methods that offer higher specificity and can minimize interference from IGF-I variants

  • Establish assay-specific reference intervals through multicenter collaboration

  • Standardize pre-analytical handling procedures including sample collection, processing, and storage conditions

The development of mass spectrometric methods has been particularly valuable for delivering more robust and accurate IGF-I measurements, as these techniques rely on precise mass determinations rather than antibody recognition . This approach also enables potential detection of pathogenic mutations through protein sequence analysis.

How should researchers design experiments to differentiate between IGF-I-specific effects and insulin-mediated effects?

Differentiating between IGF-I-specific and insulin-mediated effects presents a significant experimental challenge due to their structural similarities and overlapping signaling pathways. To address this challenge, researchers should incorporate the following design elements:

  • Dose-response analyses: IGF-I stimulates glucose transport at much lower concentrations than insulin, allowing for dose-dependent discrimination

  • Receptor-specific approaches:

    • Use receptor-specific blocking antibodies

    • Implement cell models with knockout or knockdown of either IGF1R or insulin receptor

    • Employ receptor-specific ligand analogs with modified binding preferences

  • Downstream signaling analysis: While both hormones activate PI3K and MAPK pathways, the duration, magnitude, and specific phosphorylation patterns often differ

  • Tissue-specific considerations: The relative expression levels of IGF1R versus insulin receptors vary across tissues, which can be leveraged to distinguish specific effects

When interpreting results, researchers should consider that the IGF1R and insulin receptor can form hybrid receptors with distinct signaling properties, further complicating the delineation of hormone-specific effects.

What are the optimal conditions for maintaining the stability and activity of recombinant human IGF-I in experimental settings?

Maintaining the stability and activity of recombinant human IGF-I is critical for ensuring reproducible and reliable experimental results. The following considerations should guide handling procedures:

  • Storage conditions:

    • Most commercial preparations are shipped at ambient temperature but require specific storage conditions (typically -20°C or -80°C) for long-term stability

    • Avoid repeated freeze-thaw cycles, which can lead to protein degradation and activity loss

  • Reconstitution protocols:

    • Follow manufacturer-specific reconstitution guidelines, typically using sterile buffers

    • Reconstituted IGF-I should be stored according to lot-specific recommendations, as outlined in the Certificate of Analysis

  • Experimental media considerations:

    • Cell culture media composition can significantly affect IGF-I stability

    • Serum proteins, particularly IGFBPs secreted by cells, may bind exogenously added IGF-I

    • Consider using defined serum-free media for experiments requiring precise IGF-I dosing

  • Activity verification:

    • Periodically assess the biological activity of stored IGF-I using established bioassays

    • Phosphorylation of AKT serves as a reliable indicator of IGF-I activity in cellular systems

Proper documentation of storage conditions, reconstitution protocols, and lot numbers is essential for experimental reproducibility and troubleshooting potential variations in biological responses.

What is the diagnostic utility of IGF-I measurements in growth hormone deficiency (GHD) research?

The diagnostic utility of IGF-I measurements in GHD research has been extensively investigated, with important limitations identified. Recent evidence indicates that IGF-I level has poor diagnostic accuracy as a standalone screening test for GHD . A comprehensive evaluation revealed that:

  • The mean IGF-I standard deviation (SD) was not significantly different between GHD and non-GHD groups (p = 0.23)

  • Receiver operating characteristic curve analysis demonstrated the best diagnostic accuracy at an IGF-I cutoff of -1.493 SD, with:

    • 0.685 sensitivity

    • 0.417 specificity

    • 0.25 positive predictive value

    • 0.823 negative predictive value

    • 0.517 area under the curve

These findings suggest that while IGF-I measurements have value in the broader diagnostic workup for GHD, they should not be used in isolation. Researchers investigating GHD should implement a multimodal diagnostic approach that includes:

  • Growth hormone stimulation tests using multiple secretagogues (e.g., clonidine, arginine, L-dopa)

  • Clinical evaluation of growth parameters and velocity

  • IGF-I measurements as a complementary biomarker

  • Consideration of additional factors that might influence IGF-I levels independent of GH status

This comprehensive approach provides more reliable diagnostic information than relying solely on IGF-I measurements.

How can researchers evaluate the efficacy and safety of recombinant human IGF-I in therapeutic applications?

Evaluating the efficacy and safety of recombinant human IGF-I in therapeutic applications requires comprehensive assessment protocols that address both short-term pharmacological effects and long-term safety considerations. Based on existing research, the following methodological framework is recommended:

  • Efficacy evaluation metrics:

    • For IGF deficiency (IGFD) and short stature: growth velocity, height standard deviation scores, and body composition measurements

    • For diabetes mellitus: glycemic control parameters (HbA1c, fasting glucose, glucose tolerance)

    • Biomarker responses: IGF-I levels, IGFBP profile changes, and metabolic parameters

  • Safety monitoring parameters:

    • Hypoglycemia assessment (frequency, severity, timing relative to administration)

    • Cardiovascular parameters (blood pressure, heart rate, echocardiographic measurements)

    • Cancer risk surveillance (particularly relevant given IGF-I's role in cell proliferation)

    • Specific attention to tissues with high IGF1R expression

  • Comparative safety assessment:

    • Unlike rhGH which is diabetogenic, rhIGF-I has hypoglycemic activity similar to insulin, necessitating different monitoring approaches

    • Despite distinct mechanisms, rhIGF-I and rhGH share many safety considerations since IGF-I mediates most GH actions

  • Study design considerations:

    • Long-term follow-up is essential given the growth-promoting and potential carcinogenic effects

    • Control groups should be carefully selected to match for underlying conditions

    • Dose-finding studies should precede efficacy trials to identify optimal therapeutic windows

This structured approach facilitates comprehensive evaluation of both therapeutic benefits and potential risks associated with recombinant human IGF-I intervention.

What are the most reliable biomarkers for monitoring IGF-I signaling pathway activation in clinical research?

Monitoring IGF-I signaling pathway activation in clinical research requires reliable biomarkers that accurately reflect receptor engagement and downstream signaling events. The following biomarkers have demonstrated value in assessing IGF-I pathway activation:

  • Direct IGF1R activation markers:

    • Phosphorylated IGF1R levels in accessible tissues or circulating cells

    • IGF1R/Insulin receptor hybrid receptor phosphorylation status

    • Receptor internalization and turnover rates

  • Proximal signaling components:

    • Phosphorylation status of IRS proteins (particularly IRS-1 and IRS-2)

    • Activation of PI3K, indicated by phosphatidylinositol-3,4,5-trisphosphate (PIP3) levels

    • SHC phosphorylation status

  • Downstream pathway activation:

    • Phosphorylated AKT (Ser473 and Thr308) as a primary indicator of PI3K pathway activation

    • Phosphorylated ERK1/2 reflecting MAPK pathway activation

    • mTOR pathway components (p70S6K, 4E-BP1) for protein synthesis effects

  • Functional outcome markers:

    • Glucose uptake in metabolically responsive tissues

    • Protein synthesis rates using labeled amino acid incorporation

    • Gene expression profiles of IGF-I-responsive genes

When implementing these biomarkers in clinical research, consideration must be given to tissue accessibility, the temporal dynamics of signaling events, and the potential influence of other signaling pathways. Integration of multiple biomarkers typically provides more reliable assessment of pathway activation than reliance on any single marker.

What emerging technologies are advancing our understanding of IGF-I signaling dynamics?

The field of IGF-I research continues to evolve with several emerging technologies that promise to enhance our understanding of signaling dynamics and regulatory mechanisms. Key technological advances include:

  • Single-cell analysis techniques that reveal cell-to-cell variability in IGF-I responses, providing insights into heterogeneous tissue responses

  • Advanced protein structure determination methods such as cryo-electron microscopy, which have enabled more detailed visualization of IGF1R conformational changes during activation

  • Molecular dynamics simulations that model TM associations and receptor conformational changes, revealing previously unappreciated aspects of receptor activation mechanisms

  • CRISPR-Cas9 genome editing for precise manipulation of IGF-I pathway components, enabling more sophisticated functional analyses than traditional knockout approaches

  • Mass spectrometry-based proteomics for comprehensive characterization of signaling networks and post-translational modifications

These technological advances are collectively enhancing our ability to dissect the complex regulatory mechanisms governing IGF-I signaling and its physiological impacts. Researchers entering this field should consider incorporating these advanced approaches to address persistent questions about receptor specificity, signaling dynamics, and context-dependent cellular responses.

What are the current challenges in developing predictive biomarkers for IGF-I responsiveness in clinical settings?

Despite advances in IGF-I research, significant challenges remain in developing predictive biomarkers for IGF-I responsiveness in clinical settings. Current limitations and research needs include:

  • The poor diagnostic accuracy of IGF-I as a standalone biomarker for conditions like GHD suggests that more sophisticated biomarker panels are needed

  • Variability in reference intervals across different assay platforms complicates standardization and clinical interpretation

  • Interactions between IGF-I and its binding proteins create analytical challenges that affect measurement accuracy

  • Individual genetic and physiological factors may significantly modulate IGF-I responsiveness independent of circulating levels

Future research should focus on:

  • Developing multiparametric biomarker approaches that combine IGF-I levels with additional markers

  • Implementing advanced analytical methods like mass spectrometry to enhance measurement precision

  • Identifying genetic markers that predict individual responsiveness to IGF-I

  • Establishing tissue-specific biomarkers that reflect local IGF-I activity rather than solely relying on systemic measurements

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.