The human IGF2 gene resides on chromosome 11p15.5, forming an imprinted cluster with H19 . This locus includes five promoters (p0–p4) driving differential expression across developmental stages :
The gene spans 10 exons, with promoter switching ensuring tissue- and stage-specific expression . Imprinting restricts expression predominantly to the paternal allele, regulated by CTCF binding to the H19 imprinting control region (ICR) .
CTCF binds the ICR and MAR3 regions, blocking enhancer access to IGF2 on the maternal allele .
Loss of imprinting (LOI) leads to biallelic expression, observed in Beckwith-Wiedemann syndrome and cancers .
Postnatal IGF2 suppression involves E2F3 decline. In mice, E2F3 binding to the P2 promoter decreases with age, reducing Igf2 expression by >100-fold in liver and kidney .
Human studies confirm similar E2F3-IGF2 co-regulation, with E2F3 overexpression linked to IGF2 reactivation in cancers .
Fetal Growth: IGF2 is the dominant growth factor in embryogenesis, with knockout models showing severe intrauterine growth restriction .
Tissue Maintenance: Promotes stem cell self-renewal in neural progenitor cells via insulin receptor (IR) activation .
IGF2 binds multiple receptors with distinct affinities:
Proteolytic processing generates IGF2 proforms (e.g., big-IGF2(104)), which exhibit seven-fold higher IGF-2R affinity than mature IGF2, enhancing mitogenic activity in cancers .
Beckwith-Wiedemann Syndrome: LOI at 11p15.5 causes IGF2 overexpression, leading to macrosomia and cancer predisposition .
Prostate Cancer: IGF2 underexpression correlates with treatment resistance, while miR-93-5p serves as a non-invasive biomarker .
Wilms Tumor and Hepatocellular Carcinoma: E2F3-driven IGF2 reactivation promotes tumorigenesis, independent of LOI .
IGF2 overexpression in insulinomas induces hypoglycemia via IR-A activation, mimicking insulin’s metabolic effects .
Unprocessed pro-IGF2(156) evades IGFBP inactivation and exhibits superior proliferative effects in tumors .
IGF2 is a 67-amino acid secreted peptide that serves as a major growth factor, essential for normal prenatal growth and development in humans . The IGF2 gene is located on chromosome 11 and undergoes parental imprinting, being expressed primarily from the paternal allele . Unlike mouse Igf2, which declines after birth, human IGF2 continues to be expressed in multiple adult tissues, suggesting important physiological roles throughout the lifespan .
The primary functions of IGF2 include:
Promotion of cellular growth and proliferation during embryonic development
Regulation of metabolism through interactions with insulin receptors
Modulation of tissue-specific and developmental-stage-dependent processes
The human IGF2 gene displays a more complex organization compared to its mouse homolog:
Feature | Human IGF2 | Mouse Igf2 |
---|---|---|
Chromosome location | Chromosome 11 | Chromosome 7 |
Number of exons | 10 exons | 8 exons |
Promoters | 5 promoters (P1-P5) | 4 promoters (P1-P4) |
Species-specific promoters | P1 and P2 are human-specific | - |
Adult expression | Maintained in multiple tissues | Dramatically down-regulated |
Imprinting status | Primarily imprinted, but P1 can escape imprinting in some tissues | Strictly imprinted |
Human promoters P1 and P2 are species-specific, with P2 regulating two classes of IGF2 transcripts that differ due to alternative splicing of exon 5 . The human-specific P1 promoter is particularly significant as it may explain the continued expression of IGF2 in adult human tissues and shows biallelic expression (lack of imprinting) in some contexts .
Characterizing IGF2 imprinting requires specialized techniques that distinguish between maternal and paternal allelic expression:
Polymorphism-based allele identification: Using naturally occurring polymorphisms (such as the ApaI polymorphism in the 3' untranslated region) to distinguish between maternal and paternal alleles in heterozygous individuals . This approach requires careful tissue preparation, such as removal of decidua from placental samples to avoid maternal contamination.
Methylation analysis: Bisulfite sequencing or methylation-specific PCR to analyze the methylation status of differentially methylated regions (DMRs) that control IGF2 imprinting. Different methylation patterns on maternal versus paternal chromosomes directly influence IGF2 expression.
Chromatin immunoprecipitation (ChIP): Analyzing histone modifications and transcription factor binding (particularly CTCF) at regulatory regions to understand the chromatin-level mechanisms of imprinting control.
Chromosome conformation capture: Techniques such as 3C, 4C, or Hi-C to investigate long-range chromatin interactions that establish imprinting domains and influence gene expression.
Single-cell expression analysis: New single-cell approaches can reveal cell-type specific variations in imprinting status that might be masked in bulk tissue analysis .
When designing imprinting studies, researchers must consider tissue specificity, as some human tissues (particularly those expressing IGF2 from the P1 promoter) may show biallelic expression rather than strict imprinting .
Disruptions in IGF2 regulation lead to two prominent imprinting disorders that represent molecular and phenotypic mirrors of each other :
Beckwith-Wiedemann Syndrome (BWS):
Characterized by overgrowth, macroglossia, abdominal wall defects, and increased tumor risk
Molecular causes include:
Loss of methylation at maternal ICR2/KvDMR1
Gain of methylation at maternal ICR1
Paternal uniparental disomy of chromosome 11
Results in IGF2 overexpression
Silver-Russell Syndrome (SRS):
Characterized by intrauterine and postnatal growth restriction, body asymmetry
Molecular causes include:
Hypomethylation of the paternal ICR1 leading to reduced IGF2 expression
Maternal uniparental disomy of chromosome 7 (in some cases)
Results in IGF2 underexpression
Experimental approaches to study these disorders include:
Methylation analysis of patient samples using methylation-specific PCR or bisulfite sequencing
Allele-specific expression studies to confirm altered IGF2 expression
CRISPR/Cas9-based models to recreate specific epigenetic defects
Patient-derived cell lines and organoids to study molecular mechanisms
Mouse models with targeted mutations in imprinting control regions
These disorders highlight how precise regulation of IGF2 is essential for normal growth and development, with both increased and decreased expression leading to pathological states .
Recent structural biology studies have revealed detailed mechanisms of IGF2 binding to and activation of insulin receptors (IR):
IGF2 activates insulin receptors through a distinctive mechanism compared to insulin itself. Recent cryo-EM structures of full-length human IR isoforms (IR-A and IR-B) in both inactive and IGF2-bound active states provide key insights :
Binding configuration: Under saturated IGF2 concentrations, both IR-A and IR-B adopt predominantly asymmetric conformations with two or three IGF2 molecules bound at site-1 and site-2. This differs from insulin, which induces an exclusively T-shaped symmetric conformation.
Binding affinity differences: IGF2 exhibits a relatively weak binding to IR site-2 compared to insulin, making it less potent in promoting full IR activation. This explains its differential signaling properties.
Isoform-specific interactions: In the inactive state, the C-terminus of α-CT of IR-B contacts the FnIII-2 domain of the same protomer, which hinders its threading into the C-loop of IGF2. This structural feature reduces the association rate of IGF2 with IR-B compared to IR-A .
Activation mechanism: Despite these differences, IGF2 still activates similar downstream signaling pathways through autophosphorylation of the insulin receptor tyrosine kinase domains, though with distinct signaling kinetics and potentially different cellular outcomes.
These structural insights explain the differential binding affinities of IGF2 for IR-A versus IR-B isoforms and provide a molecular basis for understanding the biological specificity of IGF2 action .
Human IGF2 gene expression is regulated by multiple transcription factors and regulatory mechanisms:
E2F transcription factors: E2F3 has been identified as a key regulator of IGF2 expression. E2f3 mRNA expression, protein levels, and binding to the Igf2 promoter decrease with age postnatally in multiple organs. Experimental restoration of E2F3 expression in juvenile hepatocytes can restore high Igf2 expression, demonstrating a causal relationship .
Promoter-specific regulation: The five human IGF2 promoters (P1-P5) are regulated by different transcription factors:
Chromatin structure: The three-dimensional organization of the IGF2-H19 locus, including enhancer-promoter interactions and boundary elements, plays a crucial role in regulating expression.
DNA methylation: Methylation patterns at differentially methylated regions (DMRs) directly influence promoter activity and accessibility to transcription factors.
Experimental approaches to study these mechanisms include:
ChIP-seq for identifying transcription factor binding sites
Reporter gene assays to test promoter activity
CRISPR/Cas9-mediated genome editing to validate regulatory elements
Chromosome conformation capture to map enhancer-promoter interactions
The complex regulation of IGF2 ensures proper temporal and spatial expression patterns during development and adult life .
IGF2 is emerging as a potential therapeutic target for Alzheimer's disease (AD) based on its neurotrophic and neuroprotective properties:
Current evidence:
IGF2 is widely expressed in the central nervous system
It functions as a crucial mechanism for synaptic plasticity, learning, and memory
Neurotrophic pathways are believed to be critical for functional recovery in AD
Current treatments that don't address neurotrophic mechanisms have limited efficacy in reversing cognitive decline
Proposed mechanisms:
IGF2 may enhance synaptic function and protect against synapse loss
It could promote neuronal survival through anti-apoptotic pathways
IGF2 might regulate neuroinflammation and microglial function
It may influence tau phosphorylation or amyloid processing
Recommended research approaches:
Comprehensive expression profiling of IGF2 and its receptors in AD brain tissues
Functional studies using IGF2 administration in multiple AD mouse models
Investigation of IGF2's effects on amyloid pathology, tau phosphorylation, and neuroinflammation
Analysis of IGF2 signaling in neurons, astrocytes, and microglia
Development of blood-brain barrier-penetrant IGF2 mimetics
Clinical correlations between IGF2 levels in cerebrospinal fluid and cognitive function
The potential of IGF2 as a therapeutic target lies in its ability to address both the structural and functional aspects of neurodegeneration in AD, potentially providing a more comprehensive approach than current treatments .
IGF2 overexpression is frequently observed in various cancers and contributes to tumor development through multiple mechanisms:
Oncogenic mechanisms:
Loss of imprinting leading to biallelic expression and increased IGF2 levels
Activation of IGF1R and IR-A signaling promoting cell proliferation and survival
Enhancement of the PI3K/AKT/mTOR pathway driving cancer cell metabolism
Promotion of angiogenesis and metastasis through downstream signaling
Creation of an autocrine growth stimulation loop
Recommended experimental approaches:
Genomic and epigenetic analysis: Examining IGF2 imprinting status, copy number alterations, and promoter methylation in tumor samples
Expression profiling: Quantifying IGF2 transcript and protein levels across cancer types and correlating with clinical outcomes
Signaling pathway investigation: Analyzing receptor activation and downstream pathways using phospho-specific antibodies and inhibitors
Functional studies: Using CRISPR/Cas9 to knock out IGF2 or its receptors in cancer cell lines and xenograft models
Preclinical therapeutics: Testing IGF2-targeting antibodies, receptor antagonists, or pathway inhibitors
Multi-omics integration: Combining transcriptomic, proteomic, and metabolomic data to understand IGF2's role in cancer cell metabolism
Cancer-specific considerations:
Understanding IGF2's role in cancer requires investigation of both genetic and epigenetic mechanisms, with particular attention to the unique aspects of IGF2 regulation in human cells that may contribute to its oncogenic potential .
Studying IGF2 receptors presents several unique challenges that require specialized approaches:
Receptor complexity and cross-reactivity:
IGF2 binds to multiple receptors (IGF1R, IR-A, IR-B, IGF2R) with different affinities
Significant homology between receptors complicates specific targeting
Hybrid receptors (IGF1R/IR) further increase complexity
Recommended approach: Use receptor-specific antibodies validated by knockout controls; employ CRISPR/Cas9 to generate receptor-deficient cell lines; use receptor-specific kinase inhibitors in combination with selective ligands.
Structural analysis challenges:
Full-length receptors are large membrane proteins difficult to study structurally
Conformational changes upon ligand binding are complex and dynamic
Different receptor isoforms may adopt distinct conformations
Recommended approach: Employ cryo-EM for full-length receptor structures as demonstrated in recent studies ; use hydrogen-deuterium exchange mass spectrometry for mapping conformational changes; combine computational modeling with experimental validation.
Signaling pathway overlap:
IGF2-activated receptors trigger multiple overlapping pathways
Temporal dynamics of signaling are critical but difficult to capture
Pathway cross-talk complicates interpretation
Recommended approach: Use phospho-proteomics to capture pathway activation comprehensively; employ live-cell imaging with fluorescent biosensors; analyze signaling at single-cell level to address heterogeneity.
Binding protein interference:
IGF binding proteins (IGFBPs) modulate IGF2 availability and activity
Standard assays may not account for binding protein effects
Tissue-specific IGFBP expression patterns alter local IGF2 activity
Recommended approach: Include analysis of IGFBPs in experimental design; use acid-ethanol extraction to release IGF2 from binding proteins; develop assays that can measure free versus bound IGF2.
Species differences:
Human and mouse receptors show differences in binding properties
Cellular contexts affect receptor expression and signaling
Model systems may not fully recapitulate human receptor biology
Recommended approach: Use human cell lines or humanized mouse models; directly compare human and mouse systems in parallel experiments; validate findings across multiple model systems.
Recent structural studies of IGF2 binding to insulin receptors demonstrate how these challenges can be overcome through integration of advanced techniques like cryo-EM with functional assays .
Analyzing IGF2 transcript variants requires specialized approaches due to the gene's complex promoter usage and tissue-specific expression patterns:
RNA sequencing considerations:
Promoter-specific primers: Design primers targeting unique 5' exons to distinguish transcripts from different promoters (P1-P5)
Long-read sequencing: Use technologies like PacBio or Oxford Nanopore to capture full-length transcripts and identify novel splice variants
Single-cell RNA-seq: Apply to detect cell-type specific expression patterns within tissues
Spatial transcriptomics: Map IGF2 transcript variants to specific tissue regions
Quantitative analysis approaches:
Absolute quantification: Use digital PCR with promoter-specific primers to determine copy numbers of each transcript variant
Relative expression: Apply RT-qPCR with internal controls optimized for developmental stages
Allele-specific expression: Design assays that can distinguish maternal from paternal transcripts
Transcript stability assessment: Measure half-lives of different transcript variants using actinomycin D chase experiments
Bioinformatic analysis pipeline:
Isoform-specific quantification: Use tools like Salmon or RSEM with custom transcript annotations
Differential expression analysis: Apply DESeq2 or EdgeR with covariates for developmental stage and tissue type
Correlation analysis: Identify genes co-regulated with specific IGF2 transcript variants
Visualizations: Generate comprehensive heatmaps and tissue-specific expression profiles
Integration with epigenetic data:
Correlate transcript variant expression with promoter methylation status
Analyze chromatin accessibility (ATAC-seq) at each promoter region
Examine histone modifications associated with active versus inactive promoters
Map long-range enhancer interactions using 4C or Hi-C data
Database resources:
GTEx for tissue-specific expression in adults
Human Developmental Biology Resource for embryonic expression
FANTOM5 for promoter usage across tissues
Custom dashboards integrating multiple data types
When analyzing and reporting IGF2 transcript data, researchers should clearly specify which promoters and transcript variants they are examining, as each may have distinct regulation and function across development .
The divergent regulation of IGF2 between human adults (continued expression) and mice (dramatic downregulation after birth) presents a fascinating evolutionary puzzle that can be investigated through multiple approaches:
Comparative genomic analysis:
Sequence comparison of IGF2 loci across primates, rodents, and other mammals to identify human-specific regulatory elements
Particular focus on the human-specific P1 promoter that escapes imprinting in some tissues
Analysis of transcription factor binding site evolution in promoter and enhancer regions
Identification of species-specific transposable elements that may have contributed to regulatory divergence
Epigenetic landscape comparison:
Cross-species comparison of DNA methylation patterns at IGF2 loci throughout development
Analysis of chromatin modifications and accessibility at promoters across species
Investigation of three-dimensional chromatin structure differences
Comparison of imprinting mechanisms and their developmental dynamics
Functional testing of species-specific elements:
Creation of reporter constructs with human versus mouse regulatory elements
CRISPR-mediated swapping of regulatory regions between species
Generation of humanized mouse models carrying the human IGF2 locus
Testing whether human-specific elements maintain expression in adult mouse tissues
Physiological significance investigation:
Analysis of metabolic and growth phenotypes associated with maintained IGF2 expression
Comparison of tissue regeneration capacity between species
Investigation of potential roles in tissue homeostasis unique to longer-lived species
Correlation with life history traits (longevity, reproductive timing, etc.)
Pathological implications:
Comparison of IGF2-associated disease susceptibility between humans and mice
Analysis of cancer incidence patterns that might relate to IGF2 expression differences
Investigation of metabolic disease vulnerability differences
Understanding the evolutionary forces that shaped these species-specific differences in IGF2 regulation may provide insights into human-specific aspects of growth, metabolism, and disease susceptibility .
IGF2's functions in the central nervous system represent an exciting frontier with significant therapeutic implications:
Synaptic plasticity and memory formation:
How does IGF2 modulate synaptic strength and plasticity at the molecular level?
Which neuronal subtypes primarily express and respond to IGF2?
What signaling pathways mediate IGF2's effects on long-term potentiation and memory consolidation?
How does IGF2 interact with other memory-related growth factors like BDNF?
Developmental roles:
What is IGF2's contribution to neurogenesis, neuronal migration, and circuit formation?
How does imprinting of IGF2 affect brain development and function?
Are there critical periods for IGF2 action in brain development?
What are the consequences of developmental IGF2 dysregulation for cognitive function?
Neuroprotective mechanisms:
Through which pathways does IGF2 promote neuronal survival?
Can IGF2 protect against specific neurodegenerative mechanisms (excitotoxicity, oxidative stress, protein aggregation)?
Does IGF2 regulate neuroinflammatory responses?
How does IGF2 influence blood-brain barrier function and integrity?
Therapeutic applications:
Can IGF2 administration or enhancement reverse cognitive deficits in Alzheimer's disease models?
What delivery methods can effectively target IGF2 to the brain?
Are there small molecule mimetics that can recapitulate IGF2's beneficial effects?
What are the potential side effects or safety concerns of IGF2-based CNS therapies?
Methodological approaches:
Use of conditional knockout models with brain region-specific IGF2 deletion
Application of optogenetic or chemogenetic approaches to manipulate IGF2-producing cells
Development of PET ligands to visualize IGF2 receptors in the living brain
Single-cell transcriptomics to map IGF2 and receptor expression across brain cell types
The investigation of IGF2 in the central nervous system may lead to novel therapeutic strategies for cognitive disorders, particularly Alzheimer's disease, by leveraging its neurotrophic and synaptic plasticity-enhancing properties .
The IGF-2 gene is an imprinted gene, meaning it is expressed only from the paternal allele. This epigenetic regulation is crucial for its function. Abnormalities in the imprinting of the IGF-2 gene are associated with several disorders, including Wilms tumor, Beckwith-Wiedemann syndrome, rhabdomyosarcoma, and Silver-Russell syndrome .
Recombinant human IGF-2 is produced using E. coli expression systems. The recombinant protein is typically purified to high levels of purity, often exceeding 97%, and is used in various research and therapeutic applications . The recombinant form retains the biological activity of the native protein and is used in studies involving cell proliferation, particularly in cancer research .
IGF-2 exerts its effects by binding to specific cell surface receptors, including the IGF-1 receptor and the insulin receptor. This binding activates intracellular signaling pathways that promote cell growth, differentiation, and survival. In particular, IGF-2 has been shown to stimulate cell proliferation in various cell types, including human breast cancer cells .
The role of IGF-2 in growth and development, as well as its involvement in various diseases, makes it a significant target for research. Understanding the regulation and function of IGF-2 can provide insights into developmental biology and the mechanisms underlying certain cancers and growth disorders .