Recombinant Human Insulin-like growth factor II (IGF2) (Active)

Shipped with Ice Packs
In Stock

Description

Expression Systems

  • HEK-293 Cells: Yields glycosylated IGF2 with structural fidelity to native human IGF2 .

  • E. coli: Produces non-glycosylated IGF2 at higher yields; retains bioactivity after refolding .

Functional Validation

  • Proliferation Assays:

    • ED50 <3 ng/mL in MCF-7 breast cancer cells .

    • Stimulates osteoblast mitogenesis via MAPK1/3 activation .

  • Metabolic Effects:

    • Suppresses hepatic glucose production at 14 ng/mL (comparable to insulin) .

    • Enhances glucose uptake in adipocytes and hepatocytes .

Developmental Roles

  • Fetal Growth: Regulates fetoplacental development under placental lactogen influence .

  • Neural Development: Promotes neural progenitor cell expansion via insulin receptor (IR) activation in the brain .

Disease Relevance

  • Neuroprotection:

    • Reduces α-synuclein toxicity in Parkinson’s disease models (IC50 ~50 ng/mL) .

    • Enhances dopaminergic synaptic function in vitro .

  • Cancer:

    • Stimulates MCF-7 proliferation via IGF1R/IR-A receptors .

    • Serum IGF2 correlates with VEGF levels in esophageal squamous cell carcinoma (ESCC) .

Receptor Binding Dynamics

  • IR Activation:

    • Binds IR-A/IR-B isoforms asymmetrically at site-1 and site-2, with lower site-2 affinity than insulin .

    • Weak binding at IR-B’s FnIII-2 domain reduces association rates .

  • IGF1R Interaction:

    • Engages IGF1R with higher affinity than IR, driving mitogenic signaling in cancers .

Metabolic Signaling

  • Glucose Homeostasis:

    • Inhibits hepatic glucose production (IC50 ~10 nM) via IR/PI3K-Akt pathways .

    • Synergizes with insulin in adipose tissue glucose uptake .

Applications in Research

ApplicationExperimental ModelKey OutcomeSource
Neurodegenerative Diseaseα-syn PFF-treated SN4741/SHSY5Y cells40–60% reduction in cytotoxicity
Cancer ProliferationMCF-7 cellsDose-dependent growth (EC50 1.5–6 ng/mL)
Metabolic StudiesPrimary hepatocytesSuppressed glucose production at 14 ng/mL

Product Specs

Buffer
Lyophilized from a 0.2 μm filtered solution containing 20 mM Glycine-HCl, 4% Sucrose, 4% Mannitol, and 0.02% Tween 80 (w/v), pH 3.0.
Form
Lyophilized powder
Lead Time
Product shipment typically occurs within 5-10 business days of order receipt. Delivery times may vary depending on shipping method and destination. Please contact your local distributor for precise delivery estimates.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to ensure contents settle. Reconstitute the protein in sterile, deionized water to a final concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and storing in aliquots at -20°C or -80°C. Our standard protocol utilizes 50% glycerol.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations maintain stability for 6 months at -20°C/-80°C, while lyophilized formulations are stable for 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C or -80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
C11orf43; IGF 2; IGF II; IGF-II; IGF2; IGF2_HUMAN; IGFII; INSIGF; insulin like growth factor 2 (somatomedin A) ; Insulin like Growth Factor 2; Insulin like growth factor II; Insulin like growth factor II precursor; Insulin like growth factor type 2; pp9974; Preptin; putative insulin like growth factor II associated protein; Somatomedin A; Somatomedin-A
Datasheet & Coa
Please contact us to get it.
Expression Region
25-91aa
Mol. Weight
8.91 kDa
Protein Length
Full Length of Mature Protein
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
E.coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function

Insulin-like growth factors (IGFs) exhibit growth-promoting activity. IGF-II is a major fetal growth hormone in mammals, playing a crucial role in fetoplacental development and is influenced by placental lactogen. It is also involved in tissue differentiation. In adults, IGF-II likely participates in glucose metabolism within adipose tissue, skeletal muscle, and liver. It acts as a ligand for integrins, essential for IGF-II signaling. Furthermore, IGF-II positively regulates the myogenic transcription factor MYOD1 by facilitating the recruitment of transcriptional coactivators, thereby controlling muscle terminal differentiation. It also inhibits myoblast differentiation and modulates metabolism by increasing mitochondrial respiration rate. Preptin, co-secreted with insulin in a glucose-dependent manner, acts as a physiological amplifier of glucose-stimulated insulin secretion and demonstrates osteogenic properties by increasing osteoblast mitogenic activity via MAPK1/3 phosphoactivation.

Gene References Into Functions

Selected Research Highlights on IGF-II and Preptin:

  1. Serum preptin levels in women decrease after menopause and correlate positively with estradiol and bone mineral density. PMID: 29134283
  2. Elevated serum levels of both IGF-II and VEGF may serve as a prognostic biomarker for esophageal cancer. PMID: 28186102
  3. IGF-II promotes cell proliferation and epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) cells. PMID: 29970663
  4. Women with polycystic ovary syndrome exhibit significantly higher serum preptin levels than controls. PMID: 29374985
  5. Infants with intrauterine growth restriction show higher serum IGF-II levels with specific genotypes. PMID: 28460554
  6. Upregulated IGF-II expression is associated with ovarian cancer. PMID: 28829218
  7. High IGF2 DMR methylation status is associated with more aggressive features of gastric cancer. PMID: 28871451
  8. IGF-II and CDKN1C expression are significantly upregulated in placentas after assisted reproductive technology, correlating with altered DNA methylation. PMID: 29277274
  9. IGF2 is a direct target gene of miR-3941, which is downregulated in acute pneumonia. PMID: 29328418
  10. Vigilin is crucial for maintaining IGF2 imprinting. PMID: 29157910
  11. Low IGF-II serum levels are associated with pancreatic cancer. PMID: 28681154
  12. IGF2 expression is rapamycin-independent in certain cell types. PMID: 29758070
  13. IGF2 rs680 polymorphism may influence endurance in athletes. PMID: 29107196
  14. Low methylation of the Igf2 gene promoter may promote ESCC development. PMID: 29207103
  15. IGF2 is a direct target gene of miR615, influencing esophageal squamous cell carcinoma cell motility. PMID: 29115555
  16. An indel variant in the IGF2 3'UTR is associated with impaired renal function. PMID: 29889555
  17. High molecular weight IGF-2 is associated with hypoglycemia in recurrent renal cell carcinoma. PMID: 24711554
  18. Epigenetic dysregulation of IGF2-DMR0 and its interaction with KLF4 impairs IGF2 gene expression in prostate cancer. PMID: 29017567
  19. HMGA1P7 sustains H19 and Igf2 overexpression by acting as a miRNA decoy. PMID: 27874091
  20. Blocking IGF2 is a potential therapeutic strategy for hepatocellular carcinoma (HCC). PMID: 29413895
  21. IGF2 expression varies among mesenchymal stem cells from different sources. PMID: 28629288
  22. IGF-2 is persistently expressed in oculomotor neurons, potentially impacting oculomotor resistance in amyotrophic lateral sclerosis (ALS). PMID: 27180807
  23. miR-663b, epigenetically repressed by HOTAIR, targets IGF2 in pancreatic cancer. PMID: 27895308
  24. IGF-II-mediated E-cadherin loss contributes to hepatomegaly and abnormal cell growth. PMID: 27486970
  25. Aberrant IGF2 imprinting enhances stemness and resistance to therapy in cancer stem cells. PMID: 27275535
  26. DNMT1-mediated IGF2 upregulation contributes to resistance to histone deacetylase inhibitors (HDIs). PMID: 27582487
  27. Human H19/Igf2 imprinting control regions can functionally replace mouse counterparts. PMID: 27621468
  28. IGF2 may exert oncogenic effects partly through miR-483-mediated DLC-1 suppression. PMID: 27366946
  29. ICR1 deletions are associated with IGF2/H19 domain hypomethylation. PMID: 27701793
  30. IGF2 regulatory region methylation patterns distinguish adrenocortical carcinomas from adenomas. PMID: 27535174
  31. IGF-II siRNA inactivates the FAK/PI3K/Akt pathway, reducing cell proliferation and oncogene expression. PMID: 27768959
  32. Folic acid intake during pregnancy impacts IGF2/H19 imprinting and 1-carbon metabolism. PMID: 28778973
  33. IGF-II produced by pericentral hepatocytes promotes hepatocyte proliferation and repair. PMID: 28653763
  34. Reducing IGF2 isoform 2 expression may be a therapeutic strategy for type 2 diabetes. PMID: 28838971
  35. H19/IGF2 domain triplication is associated with BWS or SRS. PMID: 27612309
  36. Stroma-induced IGF2 promotes colon cancer progression. PMID: 28534511
  37. IGF-2/INSR paracrine crosstalk contributes to bladder cancer angiogenesis. PMID: 28295307
  38. IGF2AS methylation is altered after preterm birth at very low birth weight (VLBW). PMID: 23840686
  39. Prenatal unhealthy diet is associated with IGF2 methylation and behavioral disorders. PMID: 27535767
  40. High IGF2 expression is associated with breast cancer. PMID: 27546618
  41. IGF2 and insulin receptor A are important for uterine leiomyoma stem cell proliferation. PMID: 28324020
  42. IGF2 overexpression is associated with cancer. PMID: 27869826
  43. Long non-coding RNA 91H influences H19/IGF2 locus epigenetic modifications in breast cancer. PMID: 27780718
  44. IGF2, via AKT1, contributes to non-canonical Wnt signaling. PMID: 26984550
  45. Elevated insulin-like growth factor 2 expression is associated with Beckwith-Wiedemann syndrome. PMID: 27650505
  46. IGF2 is an actionable epi-driver in hepatocellular carcinoma. PMID: 27614046
  47. The H19-Igf2 axis is negatively regulated by CTCF-PHB1 cooperation. PMID: 27687727
  48. miR-210 is upregulated, and IGF2 is downregulated (though not significantly) in non-obstructive azoospermia. PMID: 27535712
  49. INS-IGF2 read-through transcript is specifically expressed in pancreatic tumor tissue. PMID: 27667266
  50. IGF2 DNA methylation is unaffected in Parkinson's disease. PMID: 28081695
Database Links

HGNC: 5466

OMIM: 147470

KEGG: hsa:3481

STRING: 9606.ENSP00000391826

UniGene: Hs.272259

Involvement In Disease
Silver-Russell syndrome (SRS); Growth restriction, severe, with distinctive facies (GRDF)
Protein Families
Insulin family
Subcellular Location
Secreted.
Tissue Specificity
Expressed in heart, placenta, lung, liver, muscle, kidney, tongue, limb, eye and pancreas.

Q&A

What is Recombinant Human IGF-II and why is it important for research?

Recombinant Human IGF-II is a synthetic version of the naturally occurring Insulin-like Growth Factor II, produced through genetic engineering techniques. It is structurally homologous to proinsulin and shares approximately 70% sequence identity with IGF-I . Despite being one of the most abundant growth factors in the human body, IGF-II remains relatively understudied compared to insulin and IGF-I. For every research paper published on IGF-II, approximately three are published on IGF-I and thirty on insulin .

IGF-II is particularly important for research because it:

  • Functions as a potent mitogenic growth factor during embryonic development

  • Is expressed in multiple tissues and cell types with potential autocrine, paracrine, and endocrine functions

  • Exhibits highly conserved structure across species (100% identity between human, bovine, and porcine proteins)

  • Has complex regulation mechanisms that are dramatically altered during development and disease states

How does IGF-II differ structurally and functionally from IGF-I and insulin?

While IGF-II belongs to the insulin-like family and shares structural similarities with both insulin and IGF-I, it has several distinguishing features:

FeatureIGF-IIIGF-IInsulin
Size67 amino acids70 amino acids51 amino acids (processed form)
Sequence identity with IGF-II100%~70%Lower
Primary developmental roleEmbryonic/fetal growthBoth embryonic and postnatal growthMetabolic regulation
Postnatal expressionDramatically down-regulatedMaintained at significant levelsMaintained throughout life
Receptor bindingIGF1R, IGF2R, Insulin receptorIGF1R, Insulin receptor (lower affinity)Insulin receptor
Regulation complexityMost complexIntermediateLess complex than IGFs

IGF-II has evolved an incredibly intricate series of checks and balances to control its cellular activity, far more complex than those for IGF-I and insulin, suggesting that stringent control of IGF-II is extremely important for cellular function .

What are the primary receptors for IGF-II and how do they affect its biological activity?

IGF-II interacts with multiple receptors, each mediating different biological responses:

  • IGF-I Receptor (IGF1R):

    • IGF-II engages IGF1R to mediate embryonic growth

    • Activates intracellular signaling pathways promoting cell proliferation and survival

    • Binding affinity is lower than that of IGF-I for this receptor

  • IGF-II Receptor (IGF2R):

    • Functions primarily as a "sink" receptor leading to IGF-II degradation

    • Does not possess intrinsic tyrosine kinase activity

    • High-affinity binding involves multiple domains, particularly domain 11 with contributions from the fibronectin type II (FNII) region

  • Insulin Receptor (IR):

    • IGF-II can bind to IR, particularly the IR-A isoform

    • This interaction contributes to metabolic and mitogenic effects

The balance between these receptor interactions is critical for regulating IGF-II bioavailability and biological effects.

What are the recommended protocols for reconstitution and storage of recombinant IGF-II?

For optimal results when working with recombinant human IGF-II, follow these handling procedures:

Reconstitution Protocol:

  • Centrifuge vial before opening to collect material at the bottom

  • Gently pipette the recommended solution down the sides of the vial to avoid protein denaturation

  • DO NOT VORTEX the solution, as this can damage the protein structure

  • Allow several minutes for complete reconstitution before use

Storage Recommendations:

  • For prolonged storage, dilute to working aliquots in a 0.1% BSA solution

  • Store at -80°C in small, single-use aliquots

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

  • Working solutions may be stored at 4°C for up to 1 week

How can researchers accurately measure IGF-II binding affinities to receptors?

Surface plasmon resonance (SPR) is the gold standard method for measuring IGF-II binding affinities to its receptors. Based on published protocols:

Recommended SPR Protocol:

  • Couple IGF2R fragments (e.g., domains 10-13) to a biosensor surface via amine groups to achieve approximately 2000 resonance units

  • Conduct binding analysis at 25°C in HBS-EP buffer at a flow rate of 40 μl/min

  • Prepare IGF-II and mutants at concentrations ranging from 6.25-100 nM

  • Use a two-state conformational change model for curve fitting, as this best describes the 1:1 binding interaction with conformational change upon binding

  • Include appropriate regeneration steps (e.g., 10 mM HCl for 1.5 min or 2 M MgCl₂ for 2 min)

Alternative coupling methods include biotinylation of IGF2R constructs and immobilization to streptavidin-coated sensor chips. Note that direct amine coupling typically leads to lower absolute binding affinity measurements compared to biotinylation methods, though relative binding affinities remain consistent .

What challenges are associated with producing and purifying recombinant IGF-II?

Researchers commonly encounter several challenges when working with recombinant IGF-II:

  • Proper Folding:

    • IGF-II contains three disulfide bonds that must form correctly for biological activity

    • Expression systems require appropriate oxidizing environments and chaperones

  • Aggregation Concerns:

    • IGF-II has hydrophobic regions that can promote aggregation

    • Addition of stabilizers (e.g., BSA) is often necessary for maintaining solubility

  • Post-translational Modifications:

    • Different expression systems may yield variations in glycosylation patterns

    • Escherichia coli systems (commonly used) produce non-glycosylated IGF-II

  • Purification Challenges:

    • Separation from IGF-binding proteins that co-purify with IGF-II

    • Need for multiple chromatography steps to achieve high purity

  • Activity Verification:

    • Essential to confirm biological activity after purification

    • Typically assessed through cell proliferation assays with IGF-responsive cell lines

How can recombinant IGF-II be used to study developmental processes?

IGF-II plays critical roles in embryonic development, making recombinant IGF-II an important tool for developmental studies:

Key Experimental Approaches:

  • Ex vivo Organ Culture Systems:

    • Supplement culture media with defined concentrations of recombinant IGF-II

    • Analyze effects on organ growth, differentiation, and morphogenesis

    • Compare with IGF-I supplementation to distinguish specific developmental effects

  • Developmental Timing Studies:

    • Administer IGF-II at different developmental stages to identify critical windows of action

    • Monitor E2F3 expression in parallel, as E2F3 appears to drive IGF-II expression during development

  • Cell Lineage Specification:

    • Examine effects of IGF-II on stem cell differentiation into specific lineages

    • Use in conjunction with other growth factors to optimize differentiation protocols

  • Tissue-Specific Knockdown/Knockout Models:

    • Combine recombinant IGF-II administration with genetic models featuring IGF-II deficiency

    • Assess rescue effects to determine tissue-specific requirements

What are the best cellular assays to evaluate IGF-II biological activity?

Researchers can employ several established assays to evaluate the biological activity of recombinant IGF-II:

  • Cell Proliferation Assays:

    • MCF-7 breast cancer cells are highly responsive to IGF-II

    • Measure BrdU incorporation or use MTT/MTS colorimetric assays

    • Compare with known standards to establish relative potency

  • Receptor Activation Assays:

    • Western blot analysis of IGF1R phosphorylation

    • Downstream signaling activation (Akt, ERK1/2, p70S6K)

    • Establish dose-response curves (typically 0.1-100 ng/mL range)

  • Reporter Gene Assays:

    • Cells transfected with luciferase constructs under control of IGF-responsive elements

    • Allows quantitative measurement of transcriptional activation

  • Migration/Invasion Assays:

    • Transwell or wound healing assays to assess motility responses

    • Particularly relevant for studying IGF-II's role in cancer cell behavior

  • Specialized Functional Assays:

    • Regulatory T-cell function enhancement evaluation

    • Enzyme delivery assessments for lysosomal storage disorders

How should researchers account for endogenous IGF binding proteins in experimental systems?

IGF binding proteins (IGFBPs) significantly impact IGF-II bioavailability and activity in experimental systems:

Methodological Considerations:

  • Serum Considerations:

    • Fetal bovine serum contains variable levels of IGFBPs

    • Consider using defined serum replacement or carefully characterized serum lots

    • Pre-treatment of serum with acid-ethanol extraction can remove endogenous IGFBPs

  • Cellular Production of IGFBPs:

    • Many cell lines produce IGFBPs that can interfere with exogenous IGF-II

    • Measure IGFBP levels in conditioned media

    • Consider using IGFBP antibodies or IGF-II analogs with reduced IGFBP binding

  • Direct vs. Indirect Effects:

    • Include controls to distinguish direct IGF-II effects from IGFBP-mediated effects

    • Some cellular responses may be due to displacement of endogenous IGFs from IGFBPs

  • Quantification Methods:

    • Use free IGF-II-specific assays rather than total IGF-II measurements

    • Consider the impact of IGFBP proteases that may be present in the system

How can researchers investigate the relationship between E2F3 and IGF-II in development and cancer?

Recent evidence suggests E2F3 is a key regulator of IGF-II expression both in development and cancer . Researchers can explore this relationship through:

Investigative Approaches:

  • Temporal Expression Analysis:

    • Measure E2F3 and IGF-II expression levels across developmental stages

    • Data indicates E2f3 mRNA expression, protein expression, and binding to the Igf2 promoter all decrease with age postnatally in multiple mouse organs

  • Chromatin Immunoprecipitation (ChIP):

    • Confirm direct E2F3 binding to IGF-II promoter regions

    • Focus on the P2 promoter in mouse (equivalent to human P3), which contains consensus E2F-binding sites

  • Promoter-Reporter Assays:

    • Construct reporter systems containing the IGF-II promoter

    • Test activation by E2F3 in different cellular contexts

    • Include mutated E2F binding sites as controls

  • Cancer Models:

    • Compare E2F3 and IGF-II expression in normal vs. cancer tissues

    • Analyze correlation between E2F3 and IGF-II levels in human cancer datasets

    • Microarray data reveals that E2F3-overexpressing prostate and bladder cancers show increased IGF-II expression with positive correlation between E2F3 and IGF-II mRNA levels

  • Mechanistic Manipulation:

    • Use gain- and loss-of-function approaches for E2F3

    • In late juvenile hepatocytes, restoration of high E2f3 expression restored high Igf2 expression, supporting a causal relationship

What techniques are available for studying IGF-II signaling pathway specificity?

Understanding the signaling specificity of IGF-II is crucial for elucidating its biological functions:

Advanced Methodological Approaches:

  • Receptor-Specific Mutants and Antagonists:

    • Use IGF-II analogs with altered receptor binding specificities

    • Apply receptor-specific blocking antibodies to isolate signaling through individual receptors

  • CRISPR/Cas9 Receptor Modification:

    • Generate cell lines with specific receptor knockouts or mutations

    • Create cells expressing receptor variants with altered downstream signaling capabilities

  • Phosphoproteomics:

    • Employ mass spectrometry-based approaches to identify phosphorylation events

    • Compare IGF-II-induced phosphorylation profiles with those of IGF-I and insulin

    • Temporal analysis can reveal signaling dynamics and feedback mechanisms

  • Single-Cell Analysis:

    • Investigate heterogeneity in IGF-II responses within seemingly homogeneous populations

    • Correlate receptor expression levels with signaling outputs at single-cell resolution

  • Biosensors and Live Imaging:

    • Utilize FRET-based biosensors to monitor signaling events in real-time

    • Track receptor internalization and trafficking using fluorescently tagged IGF-II

How does the IGF-II/IGF2R interaction structure inform the development of targeted therapeutics?

The complex structural relationship between IGF-II and IGF2R provides opportunities for developing targeted therapeutics:

Structure-Based Design Considerations:

  • Critical Binding Domains:

    • Domain 11 of IGF2R is primarily responsible for IGF-II binding

    • The fibronectin type II (FNII) domain indirectly enhances binding affinity by interacting with the AB-loop of domain 11

  • Key Residue Interactions:

    • The Glu1544Lys mutation in isolated domain 11 increases IGF-II affinity by sixfold

    • This mutation may enhance affinity by introducing an electrostatically favorable interaction with Asp20 of IGF-II

  • Binding Affinity Modulation:

    • Deletion of FNII markedly reduces IGF-II binding, causing a 10-fold drop in binding affinity

    • The Glu1544Lys mutation has different effects depending on the context:

      • Negative impact in IGF2R-Dom10-13 (10-fold reduction in affinity)

      • Positive impact in isolated domain 11

      • Increases affinity fourfold compared with ΔFNII alone when FNII is absent

  • Therapeutic Design Strategies:

    • Design peptides targeting the IGF-II binding interface of domain 11

    • Develop antibodies that enhance IGF2R-mediated IGF-II clearance

    • Create IGF-II variants with altered receptor specificity profiles for targeted applications

How is IGF-II dysregulation implicated in cancer development and progression?

IGF-II dysregulation plays significant roles in multiple cancer types:

Key Research Findings:

  • Overexpression Patterns:

    • IGF-II is frequently overexpressed in childhood malignancies including Wilms tumor, medulloblastoma, and rhabdomyosarcoma

    • Also commonly overexpressed in adult cancers: lung, breast, colorectal, bladder, ovarian, and liver cancers

  • Mechanisms of Overexpression:

    • Loss of imprinting (LOI) where the normally silenced maternal IGF-II allele becomes expressed

    • This biallelic expression typically leads to approximately doubled expression

    • Many cancers show much higher increases in IGF-II, suggesting additional mechanisms

    • Aberrant activation of the P3 promoter (human, equivalent to mouse P2) is observed in multiple cancers

    • E2F3 overexpression appears to drive IGF-II overexpression in some cancers, particularly prostate and bladder cancers

  • Functional Consequences:

    • Promotes cancer cell proliferation, survival, and invasion

    • Contributes to tumor angiogenesis

    • May facilitate metastatic spread

  • Research Approaches:

    • Analyze correlations between E2F3 and IGF-II expression in cancer databases

    • Investigate promoter usage patterns in different cancer types

    • Examine effects of IGF-II neutralization on cancer cell behavior

What experimental models are best suited for studying IGF-II in specific disease contexts?

Researchers investigating IGF-II in disease states can utilize various experimental models:

Model Systems for IGF-II Research:

  • Cell Culture Models:

    • Cancer cell lines with variable IGF-II expression

    • Primary cells from disease-relevant tissues

    • 3D organoid cultures to better recapitulate tissue architecture

  • Genetic Mouse Models:

    • Igf2 knockout or conditional knockout mice

    • Transgenic mice with tissue-specific IGF-II overexpression

    • Humanized IGF-II mice to better model human-specific regulation

    • E2f3 manipulation models to study regulation of IGF-II expression

  • Patient-Derived Xenografts (PDX):

    • Capture tumor heterogeneity and microenvironment interactions

    • Allow testing of IGF-II-targeting approaches in clinically relevant models

  • Disease-Specific Models:

    • For cancer: Orthotopic implantation models for tissue-specific microenvironments

    • For metabolic disorders: Diet-induced models with IGF-II pathway modulation

    • For developmental disorders: Ex vivo organ culture systems with recombinant IGF-II supplementation

  • Disease Applications Beyond Cancer:

    • Mucopolysaccharidosis type IIIB models to study IGF-II peptide fusion for enzyme delivery

    • Food allergy models to investigate IGF-II's role in regulatory T-cell functions

How can researchers develop IGF-II-based therapeutic approaches?

IGF-II's diverse biological functions offer several avenues for therapeutic development:

Therapeutic Development Strategies:

  • IGF-II as a Therapeutic Target:

    • Neutralizing antibodies against IGF-II

    • Antisense oligonucleotides or siRNAs to reduce IGF-II expression

    • Small molecule inhibitors of IGF-II/receptor interactions

    • Targeting E2F3 to modulate IGF-II expression in cancers

  • IGF-II as a Therapeutic Agent:

    • Recombinant IGF-II for conditions requiring growth promotion

    • IGF-II fusion proteins for targeted delivery applications

    • IGF-II peptide fusion enables uptake and lysosomal delivery of α-N-acetylglucosaminidase to mucopolysaccharidosis type IIIB fibroblasts

  • Receptor Targeting Approaches:

    • IGF2R-targeted therapies to enhance IGF-II clearance

    • Selective targeting of IGF1R vs. insulin receptor signaling

    • Bispecific antibodies engaging multiple components of the IGF system

  • Methodological Considerations:

    • Cell-based screening assays for candidate molecule identification

    • Structure-based design leveraging the IGF-II/IGF2R interaction data

    • Appropriate animal models for efficacy and safety assessment

    • Consideration of compensatory mechanisms in the insulin/IGF system

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.