IGF2 Mouse Recombinant produced in E.Coli is a single, non-glycosylated, monomeric polypeptide chain containing 67 amino acids and having a total molecular mass of 7.4kDa.
The IGF2 is purified by proprietary chromatographic techniques.
Insulin-like growth factor II (IGF-II) is a crucial hormone for fetal growth, primarily produced by theca cells during pregnancy. It plays a dual role by binding to the IGF-II receptor (IGF2R), leading to its degradation, and interacting with the IGF-I receptor (IGF1R) to regulate embryonic development.
Recombinant Mouse IGF2, expressed in E. coli, is a single-chain polypeptide consisting of 67 amino acids. This non-glycosylated, monomeric protein has a molecular weight of 7.4kDa. The purification process involves proprietary chromatographic techniques.
Lyophilized from a sterile (0.2µm) filtered solution containing 0.1% trifluoroacetic acid (TFA).
For reconstitution, it is recommended to dissolve the lyophilized IGF2 in sterile water to achieve a concentration of 0.1 mg/ml. This solution can be further diluted with other aqueous solutions as needed.
Lyophilized IGF2 remains stable at room temperature for up to 3 weeks; however, for long-term storage, it is recommended to store it desiccated below -18°C. After reconstitution, the IGF2 should be stored at 4°C for 2-7 days. For extended storage, freezing below -18°C is advised, preferably with the addition of a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
The ED50, determined by FDC-P1 cell proliferation assay, is ≤ 50 ng/mL. This corresponds to a specific activity of ≥ 2.0 x 104 units/mg.
Somatomedin-A, IGF2, INSIGF, pp9974, C11orf43, FLJ22066, FLJ44734
Escherichia Coli.
AYGPGETLCG GELVDTLQFV CSDRGFYFSR PSSRANRRSR GIVEECCFRS CDLALLETYC ATPAKSE
The mouse Igf2 gene resides on chromosome 7 within a conserved linkage group that mirrors the human IGF2 organization on chromosome 11p15.5 . Genomic analysis reveals a complex structure with multiple 5' non-coding exons linked to distinct promoters, allowing tissue-specific and developmental stage-dependent regulation . The coding exons of Igf2 show high conservation across mammalian species, reflecting the essential function of this growth factor. Research techniques to characterize this organization include genomic DNA sequencing, RNA-seq analysis of different tissues, and promoter mapping using 5' RACE (Rapid Amplification of cDNA Ends) . The gene's architecture features multiple regulatory elements, including a putative imprinting control region 5' to the neighboring H19 gene and potential enhancer elements 3' to H19 .
Igf2 is a parentally imprinted gene in mice, with expression predominantly from the paternally derived chromosome . This imprinting is established through differential DNA methylation at imprinting control regions (ICRs). Research approaches to study imprinting include bisulfite sequencing to map methylation patterns, chromatin immunoprecipitation to analyze histone modifications, and allele-specific expression analysis using polymorphic markers . In experimental studies, researchers can distinguish between maternal and paternal alleles using polymorphisms in the gene's 3' untranslated region, as demonstrated in placental studies where only the paternal allele produces mRNA . Cross-breeding strategies with polymorphic mouse strains are essential for tracking parent-of-origin expression patterns.
IGF2 functions as a major fetal growth factor in mice. Targeted disruption of Igf2 results in severe growth retardation at birth, demonstrating its critical role in normal somatic development . The expression of Igf2 is highest during embryonic and fetal development, followed by dramatic down-regulation in multiple organs after birth . This temporal expression pattern correlates with its primary functions in promoting cell proliferation, survival, and differentiation during organogenesis. Recent studies have also revealed unexpected roles in neurodevelopment and behavior, as IGF2 administration can rescue social deficits in mouse models of autism spectrum disorders .
Research has identified E2F transcription factor 3 (E2F3) as a key regulator of postnatal Igf2 down-regulation . E2f3 mRNA expression, protein expression, and binding to the Igf2 promoter all decrease with age postnatally in multiple mouse organs . Experimental approaches to study this regulation include chromatin immunoprecipitation to assess transcription factor binding, reporter gene assays to test promoter activity, and restoration experiments in juvenile cells. Critically, restoration of high E2f3 expression in late juvenile hepatocytes restored high Igf2 expression, establishing a causal relationship . This relationship was not observed in fetal hepatocytes, which already express high levels of both E2f3 and Igf2, suggesting developmental stage-specific regulatory mechanisms.
Mouse models overexpressing IGF2 show significant metabolic alterations. These mice display increased weight during early adulthood (5-10 weeks) along with reduced glucose concentration, total cholesterol, and high-density lipoprotein cholesterol (HDL-C) levels compared to wild-type controls . The metabolic effects of IGF2 overexpression are mediated through tissue-specific changes in key signaling components, as summarized in Table 1 below . These findings suggest that IGF2 plays important roles in metabolic regulation beyond its established growth functions.
Tissue | Gene Expression Changes | Metabolic Effects |
---|---|---|
Liver | ↓ STAT3, ↑ GLUT2 | Lower glucose, total cholesterol, and HDL-C |
Muscle | ↓ IGF1R, ↓ STAT3, ↑ AKT2, ↑ GLUT2 | Enhanced glucose uptake |
Kidney | ↑ GLUT2 | Altered glucose handling |
Pancreas | ↓ H19, ↓ GLUT2 | Modified insulin secretion |
While the mature IGF2 protein sequence is highly conserved across mammals, important differences exist between mouse and human IGF2 biology . Species-specific alternative RNA splicing produces protein versions of 67 or 70 residues in different mammals . Research comparing human and mouse IGF2/Igf2 has revealed differences in regulatory elements, tissue-specific expression patterns, and developmental timing of expression changes . Methodological approaches to study these differences include comparative genomics, cross-species analysis of expression patterns, and functional testing of human regulatory elements in transgenic mice. These interspecies differences must be considered when extrapolating findings from mouse models to human conditions.
Contemporary approaches for generating Igf2 mouse models rely heavily on CRISPR/Cas9-based genome editing. For overexpression models, researchers have successfully employed site-directed insertion of the Igf2 gene with loxP sites into permissive loci (such as H11 or Rosa26) . Tissue-specific expression can be achieved by crossing these mice with strains expressing Cre recombinase under tissue-specific promoters, such as the human CYP19A1 promoter for broad IGF2 overexpression . For knockout models, conditional systems are preferred to bypass embryonic lethality. Validation of these models requires comprehensive molecular characterization through genomic PCR, RNA and protein expression analysis, and phenotypic assessment . When studying imprinting effects, researchers must carefully consider the parental origin of the modified allele.
Experimental design for neurodevelopmental studies of Igf2 requires a multidisciplinary approach. Molecular techniques should include analysis of Igf2 expression in specific brain regions using in situ hybridization or region-specific RNA-seq. Cellular approaches should incorporate electrophysiology to assess synaptic function, particularly in regions implicated in social behavior such as the CA2 hippocampal region . Behavioral testing should combine social interaction paradigms (e.g., three-chamber test), cognitive assessments, and general behavioral characterization. A recent study demonstrated that systemic IGF-2 treatment could reverse social deficits in Neuroligin 3 knockout mice (NLG3–/y), a model of autism spectrum disorders, suggesting therapeutic potential . Control experiments must account for potential metabolic effects of Igf2 manipulation that might indirectly affect behavior.
When investigating metabolic phenotypes in Igf2-modified mice, comprehensive controls and validation are critical. Experimental designs should include age-matched and sex-matched wild-type controls, with monitoring of body weight and composition throughout development . Metabolic assessment should include glucose tolerance tests, insulin sensitivity assays, and comprehensive blood chemistry panels . Tissue-specific effects require analysis of gene expression in multiple organs to identify direct versus secondary consequences. Validation should include assessment of IGF2 protein levels in circulation and tissues, as well as activation status of downstream signaling pathways. Time-course studies are essential as metabolic phenotypes may evolve throughout development and aging.
Resolving conflicting results requires systematic analysis of methodological differences. For contradictory findings across studies, researchers should compare: (1) Genetic background of mouse models; (2) Age and sex of experimental animals; (3) Environmental conditions including diet and housing; (4) Specific tissues examined; (5) Analytical techniques employed; and (6) Statistical approaches used. For example, the effects of IGF2 on glucose metabolism may differ between developmental stages or genetic backgrounds . Reproducibility issues can be addressed through standardized protocols, appropriate sample sizes based on power analysis, and independent validation in multiple laboratories. Meta-analysis approaches can help identify consistent effects across studies versus context-dependent phenomena.
Developmental studies of Igf2 expression require statistical approaches that account for temporal dynamics and tissue specificity. Appropriate methods include: (1) Time-series analysis using mixed-effects models for longitudinal sampling; (2) Developmental trajectory modeling to identify co-regulated gene networks; (3) Multi-omics statistical frameworks to integrate transcriptomic, proteomic, and metabolomic data; and (4) Careful normalization strategies considering developmental changes in reference genes. Power analysis should determine appropriate sample sizes for detecting age-dependent changes. Non-parametric approaches may be necessary when data don't meet normality assumptions, which is common in developmental studies with variable growth rates.
Distinguishing direct from indirect effects of Igf2 manipulation requires complementary approaches: (1) Acute versus chronic manipulations using inducible systems; (2) Rescue experiments to restore specific downstream pathways; (3) Ex vivo studies with isolated tissues or cells to minimize systemic effects; (4) Molecular dissection of signaling pathways using phospho-specific antibodies and pathway inhibitors; and (5) Cell type-specific manipulations using Cre-loxP systems. In the case of IGF2 overexpression, observed changes in glucose metabolism may result directly from IGF2 signaling or indirectly through altered expression of metabolic regulators like GLUT2 . Temporal analysis and pathway inhibitor studies can help establish causality.
Mouse models of Igf2 dysregulation provide valuable insights into human imprinting disorders when analyzed appropriately . Methodological considerations include comparative analysis of molecular phenotypes between mouse models and patient samples, targeted engineering of human-specific regulatory mutations in mice, and comprehensive phenotyping addressing both common features (growth) and syndrome-specific manifestations. Beckwith-Wiedemann syndrome (overgrowth) and Silver-Russell syndrome (growth restriction) represent reciprocal disorders involving IGF2 dysregulation . Mouse models with Igf2 overexpression or deficiency recapitulate key aspects of these conditions, though species differences in placentation and imprinting mechanisms must be considered when translating findings to human disease.
Recent mouse studies suggest several potential therapeutic applications of IGF2 modulation. For neurodevelopmental disorders, systemic IGF-2 administration reversed social deficits in a mouse model of autism spectrum disorders (NLG3–/y), suggesting potential for treating social impairments . In metabolic research, understanding how IGF2 overexpression affects glucose and lipid metabolism could inform approaches to metabolic disorders . From a methodological perspective, therapeutic studies should include dose-response assessments, multiple administration routes, pharmacokinetic analysis, long-term safety evaluation, and comprehensive behavioral or metabolic outcome measures depending on the target condition. Translational considerations must account for species differences in IGF2 biology and potential off-target effects.
Assessing translational relevance requires systematic comparison of mouse and human IGF2 biology . Research has revealed both conservation and divergence: the mature IGF2 protein shows high sequence conservation, and imprinting mechanisms are preserved across species, but regulatory elements, expression patterns, and developmental timing show notable differences . The human and mouse IGF2/Igf2 genes each reside within conserved linkage groups on human chromosome 11p15.5 and mouse chromosome 7, respectively . Approaches to enhance translational relevance include humanized mouse models carrying human IGF2/H19 loci and validation in human cell and organoid systems. Despite differences, mouse models have successfully predicted many aspects of human IGF2 biology, particularly regarding growth phenotypes and imprinting mechanisms.
Insulin-like growth factor-2 (IGF-2) is a member of the insulin family of polypeptide growth factors, which play a crucial role in development and growth. IGF-2 is particularly significant during fetal and prenatal development, where it mediates growth-promoting activities . This protein is encoded by the IGF2 gene, which is an imprinted gene expressed predominantly from the paternal allele .
IGF-2 is a potent mitogenic growth factor that binds with high affinity to the IGF-2 receptor. It shares structural similarities with insulin but has a much higher growth-promoting activity . The IGF system, which includes IGF-1 and IGF-2, is involved in various physiological functions, including cell proliferation, differentiation, and survival .
The expression of IGF-2 is regulated by genetic and epigenetic factors. Parental imprinting plays a significant role in its regulation, with the gene being expressed only from the paternal allele . Epigenetic changes at the IGF2 locus are associated with several disorders, including Wilms tumor, Beckwith-Wiedemann syndrome, rhabdomyosarcoma, and Silver-Russell syndrome . These conditions highlight the importance of proper IGF-2 regulation for normal growth and development.
Recombinant IGF-2, such as mouse recombinant IGF-2, is produced using recombinant DNA technology. This involves inserting the IGF2 gene into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. Recombinant IGF-2 is used in various research applications to study its role in growth and development, as well as its potential therapeutic uses .
Recombinant IGF-2 is widely used in research to understand its physiological roles and regulatory mechanisms. Studies have shown that IGF-2 is involved in tissue-specific and developmental-stage-dependent actions . It is also used to investigate the effects of IGF-2 on metabolism, tumor susceptibility, and other body functions . The availability of recombinant IGF-2 has facilitated the study of its function in various model systems, including mouse models.