IGFBP6 Mouse

Insulin Like Growth Factor Binding Protein-6 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Functional Characteristics

IGFBP6 is a 213-amino-acid O-glycosylated protein with a high affinity for IGF-II (Ki ≈ 0.1–1 nM) . Mouse IGFBP6 shares 73% amino acid similarity with human IGFBP6 and 94% similarity with rat IGFBP6, with a 9-amino-acid insertion distinguishing it from rat homologs . Key structural features include conserved cysteine residues in terminal regions, critical for IGF binding and post-translational modifications (e.g., phosphorylation, proteolysis) .

FeatureDetail
Precursor Length238 amino acids (including 25-aa signal peptide)
Mature Protein213 amino acids
GlycosylationO-linked glycans enhance stability and cell-specific interactions
Tissue ExpressionOvary, testis, muscle, heart, lung; absent in whole embryo RNA

Physiological Roles in Mice

IGFBP6 exerts multifaceted effects on tissue homeostasis and disease progression.

Immune Regulation

  • Neutrophil Activation: Stimulates oxidative burst, degranulation, and chemotaxis via IGF-independent mechanisms .

  • Anti-inflammatory Effects: Reduces pro-inflammatory cytokines (e.g., TNF-α, IL-1β) in cystic fibrosis models, though its expression is elevated in F508del-CFTR bronchial epithelial cells .

Fibrosis Modulation

IGFBP6 regulates fibrotic pathways in diverse tissues:

TissueMechanismReferences
SkinDermal fibrosis inhibition via suppression of fibroblast proliferation
KidneyRenal fibrosis mitigation by modulating TGF-β signaling
LiverHepatic fibrosis attenuation through IGF-II sequestration
HeartCardiac fibrosis reduction by limiting IGF-II-driven fibroblast activation
Bone MarrowMyelofibrosis progression via SHH/TLR4 axis activation in stromal cells

Vascular Protection

Endothelial IGFBP6 suppresses atherosclerosis by:

  1. Inhibiting Inflammatory Signaling: Reduces NF-κB/JNK activation via MVP (major vault protein) interaction .

  2. Monocyte Adhesion Blockade: Overexpression reverses TNF-α-induced adhesion in endothelial cells .

CNS Development

Transgenic mice overexpressing human IGFBP6 under the GFAP promoter show:

  • Reduced Cerebellum Size: 25% smaller volume and 35% lower weight .

  • Astrocyte Differentiation Defects: Fewer GFAP+ astrocytes, implying disrupted CNS maturation .

CNS-Focused IGFBP6 Overexpression

PhenotypeObservationReference
Body WeightSignificant reduction due to lower plasma IGF-I levels
Reproductive DysfunctionSmaller litter sizes, fewer corpora lutea, and reduced LH levels

Fibrosis and Atherosclerosis

  • Myelofibrosis: IGFBP6 upregulation in primary myelofibrosis patients correlates with SHH pathway activation in stromal cells .

  • Atherosclerosis: Endothelial IGFBP6-deficient mice exhibit aggravated plaque formation, while overexpression protects against diet-induced lesions .

Recombinant IGFBP6 Protein Applications

Recombinant mouse IGFBP6 (R&D Systems, Cat. #776-B6) is used in:

Functional Studies

  • Enzyme Assays: Measured ED₅₀ of 0.08–0.96 µg/mL in inhibiting IGF-II-induced cell proliferation .

  • ELISA Standards: Carrier-free (BSA-free) formulations avoid interference in sensitive assays .

FormulationDescriptionApplication
Lyophilized (Acetonitrile/TFA)Reconstituted at 200 µg/mL in PBS; stable at -20°C Cell culture, biochemical assays
Carrier-FreeBSA-free to prevent cross-reactivity in immunoprecipitation ELISA, affinity chromatography

Therapeutic Implications

  1. Fibrotic Diseases: Targeting IGFBP6 in myelofibrosis (e.g., SHH/TLR4 inhibitors) or upregulating it in atherosclerosis .

  2. Neurological Disorders: Modulating IGFBP6 in Parkinson’s disease models, where its secretion by dental stem cells improves motor function .

  3. Cystic Fibrosis: Leveraging IGFBP6’s anti-inflammatory properties to mitigate airway inflammation .

Product Specs

Introduction
IGFBP6 is involved in lipoprotein assembly and the absorption of cholesterol from food. Besides its acyltransferase function, it might also act as a ligase. It may supply cholesteryl esters needed for lipoprotein secretion from liver cells and the lining of the gut.
Description
Recombinant Mouse IGFBP6, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It consists of 221 amino acids (26-238 a.a.), with a molecular weight of 23.7kDa (observed as 28-40kDa on SDS-PAGE under reducing conditions). This IGFBP6 protein is expressed with an 8 amino acid His tag at its C-terminus and purified using proprietary chromatographic methods.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The IGFBP6 protein solution (0.25mg/ml) is formulated in Phosphate Buffered Saline at pH 7.4 and contains 40% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For longer periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freezing and thawing cycles.
Purity
Purity exceeds 90.0% as determined by SDS-PAGE analysis.
Synonyms
Insulin-like growth factor-binding protein 6, Igfbp6, IGFBP-6, IBP-6, IGF-binding protein 6, IGFBP-6, Igfbp-6.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
ALAGCPGCGA GMQTGCRGGC VEEEDAGSPA DGCTEAGGCL RREGQPCGVY SPKCAPGLQC QPRENEEAPL RALLIGQGRC QRARGPSEET TKESKPQGGA SRSRDTNHRD RQKNPRTSAA PIRPNPVQDS EMGPCRRHLD SVLQQLQTEV FRGGARGLYV PNCDLRGFYR KQQCRSSQGN RRGPCWCVDP MGQPLPVSPD GQGSTQCSAR SSGLEHHHHH H.

Q&A

What is the basic functional significance of IGFBP6 in mice?

IGFBP6 is one of six high-affinity insulin-like growth factor binding proteins that regulate IGF actions. It is distinctive for its approximately 50-fold binding preference for IGF-II over IGF-I, making it a relatively selective inhibitor of IGF-II actions including proliferation, survival, and differentiation across various cell types . Beyond IGF regulation, IGFBP6 demonstrates multiple IGF-independent functions including promotion of apoptosis in certain cell types, inhibition of angiogenesis, and induction of cell migration particularly in tumor cells . In mouse models, IGFBP6 has been shown to influence central nervous system development, body weight regulation, and reproductive physiology .

How does mouse IGFBP6 compare structurally to human IGFBP6?

Mouse IGFBP6 shares 70-85% sequence identity with human IGFBP6, which indicates high evolutionary conservation . This conservation extends to key functional domains including:

  • The N-domain containing essential residues for IGF binding

  • The C-domain containing six conserved cysteines and the CWCV motif

  • The linker domain that undergoes post-translational modifications

The high degree of homology suggests that mouse models are suitable for studying IGFBP6 functions relevant to human biology, although species-specific differences should be considered when translating findings . Both human and mouse IGFBP6 retain critical amino acids involved in IGF binding, suggesting comparable binding affinities across species.

What is the expression pattern of IGFBP6 across mouse tissues during development and adulthood?

IGFBP6 shows a distinctive spatiotemporal expression pattern across mouse tissues. Based on comprehensive expression data, IGFBP6 is expressed in multiple tissues including:

  • Central nervous system (particularly in astrocytes)

  • Cardiovascular system

  • Reproductive system

  • Musculoskeletal system

  • Endocrine tissues

During embryonic development, IGFBP6 expression is regulated by the Hedgehog (Hh) pathway, which is critical for developmental processes . In adults, IGFBP6 expression varies by tissue type and can be modulated by physiological stressors including hypoxia, which increases IGFBP6 expression via HIF-1α . Expression data from the Mouse Genome Database indicates IGFBP6 presence across multiple organ systems, suggesting its broad physiological significance .

What are the available transgenic mouse models for studying IGFBP6 function?

Several transgenic mouse models have been developed to study IGFBP6 function in vivo:

  • GFAP-promoter driven hIGFBP6 transgenic mice: These mice express human IGFBP6 cDNA under the control of the glial fibrillary acidic protein (GFAP) promoter, targeting expression primarily to astrocytes in the central nervous system. These mice exhibit reduced cerebellum size (25% reduction) and weight (35% reduction), fewer differentiated GFAP-expressing astrocytes, and reproductive abnormalities .

  • IGFBP6-deficient mice: Complete knockout models show aggravated diet-induced atherosclerosis and disturbed flow-induced atherosclerosis, confirming IGFBP6's protective role against vascular inflammation .

  • Endothelial-specific IGFBP6-overexpressing mice: These models demonstrate protection against atherosclerosis, providing evidence for tissue-specific IGFBP6 functions .

Each model offers distinct advantages for investigating tissue-specific roles of IGFBP6, with the choice depending on the specific research question being addressed.

What methods are most effective for measuring IGFBP6 expression and activity in mouse tissues?

Multiple complementary methods can be employed to effectively measure IGFBP6 expression and activity:

For expression analysis:

  • qRT-PCR for mRNA quantification

  • Western blotting for protein levels using specific antibodies

  • Immunohistochemistry for spatial localization in tissues

  • RNA sequencing for transcriptome-wide contextual analysis

For functional activity assessment:

  • IGF binding assays to determine binding affinity for IGF-I versus IGF-II

  • Cell proliferation assays in the presence of recombinant IGFBP6

  • Migration assays to assess IGF-independent chemotactic effects

  • Angiogenesis assays (tube formation) to evaluate anti-angiogenic properties

For comprehensive characterization, researchers should combine multiple techniques. For example, Western blotting verified transgene expression in the CNS of GFAP-promoter driven hIGFBP6 mice, while histological examination revealed phenotypic consequences . When measuring IGFBP6 in circulation, it's important to note that glycosylation can affect antibody detection, potentially requiring specific antibodies that recognize post-translationally modified forms .

How can I effectively manipulate IGFBP6 expression in specific mouse tissues for functional studies?

Several approaches enable tissue-specific manipulation of IGFBP6 expression:

For overexpression:

  • Tissue-specific promoter-driven transgenes: Using promoters like GFAP for CNS targeting or Tie2 for endothelial-specific expression. This approach was successful in studying IGFBP6 function in astrocytes .

  • Viral vector delivery: Adeno-associated viruses (AAVs) with tissue-specific promoters can deliver IGFBP6 to target tissues in adult mice, avoiding developmental compensation.

  • Inducible expression systems: Tet-On/Off systems allow temporal control of IGFBP6 expression in specific tissues.

For knockdown/knockout:

  • Conditional knockout using Cre-loxP system: Enables tissue-specific deletion of IGFBP6 when crossed with appropriate Cre-driver lines.

  • siRNA delivery: Local delivery of siRNAs targeting IGFBP6 can achieve transient knockdown in specific tissues, similar to techniques used in human endothelial cells where IGFBP6 reduction increased inflammatory molecule expression .

  • CRISPR/Cas9 with tissue-specific delivery: Enables targeted genomic editing in specific cell types.

The choice of method depends on the research question, with consideration of whether constitutive or inducible modulation is needed, and whether the focus is on developmental or adult phenotypes.

Through which signaling pathways does IGFBP6 exert its biological effects in mice?

IGFBP6 functions through multiple signaling pathways that can be categorized as IGF-dependent and IGF-independent mechanisms:

IGF-dependent pathways:

  • Inhibition of IGF-II binding to IGF receptors, which prevents activation of PI3K/Akt and MAPK pathways that normally promote cell proliferation and survival .

IGF-independent pathways:

  • MAP kinase pathway: IGFBP6 induces migration of tumor cells including rhabdomyosarcomas through MAP kinase activation .

  • Prohibitin-2 interaction: IGFBP6 binds to prohibitin-2 on the cell surface to mediate cell migration through a mechanism independent of MAP kinases .

  • MVP-JNK/NF-κB signaling axis: In endothelial cells, IGFBP6 exerts anti-inflammatory effects through the major vault protein (MVP)–c-Jun N-terminal kinase (JNK)/nuclear factor kappa B (NF-κB) signaling axis .

  • Nuclear actions: IGFBP6 can enter the nucleus via a C-domain nuclear localization sequence that interacts with importin-α, potentially modulating transcription and DNA repair through interaction with proteins like Ku80 .

  • Interaction with LIM mineralization protein-1 (LMP-1): In osteoblasts, IGFBP6 inhibits differentiation by binding LMP-1 and affecting its nuclear-cytoskeletal shuttling .

The relative contribution of these pathways appears to be cell type-specific and context-dependent, requiring targeted experimental approaches to delineate in specific tissues.

How does IGFBP6 interact with other components of the IGF system in mouse models?

IGFBP6 interacts with multiple components of the IGF system in mice in a complex regulatory network:

  • Differential binding to IGF-I versus IGF-II: IGFBP6 has approximately 50-fold higher binding affinity for IGF-II compared to IGF-I, making it a relatively selective inhibitor of IGF-II actions . This contrasts with other IGFBPs that bind both ligands with similar affinities.

  • Impact on circulating IGF levels: Transgenic mice overexpressing IGFBP6 showed reduced plasma IGF-I levels between birth and 1 month of age, suggesting systemic effects on IGF regulation .

  • Proteolytic regulation: IGFBP6 is subject to specific limited proteolysis by several proteases including cathepsin-D-like acid proteases, neutral serine proteases, and matrix metalloproteinases (MMPs) such as MMP-7, MMP-2, MMP-9, and MMP-12 . This proteolysis can release bound IGF-II for receptor binding.

  • Post-translational modifications: O-glycosylation of IGFBP6 within its linker domain does not affect IGF binding but modulates protein stability and localization, influencing its regulatory capacity .

  • Compensatory expression patterns: In some cancer models, decreased IGFBP6 expression suggests a role as a tumor suppressor, while increased expression in other cancers may reflect a compensatory mechanism to control IGF-II actions .

These interactions highlight the complexity of IGFBP6 function within the broader IGF system, with consequences for development, metabolism, and pathological processes.

What is the role of IGFBP6 in inflammation and atherosclerosis based on mouse models?

Recent research has established IGFBP6 as a critical regulator of vascular inflammation and atherosclerosis in mouse models:

  • Anti-inflammatory effects: IGFBP6 functions as a homeostasis-associated molecule that restrains endothelial inflammation. IGFBP6-deficient mice showed aggravated diet-induced and disturbed flow-induced atherosclerosis, while endothelial-specific IGFBP6-overexpressing mice were protected against atherosclerosis .

  • Molecular mechanism: IGFBP6 executes its anti-inflammatory effects through the major vault protein (MVP)–c-Jun N-terminal kinase (JNK)/nuclear factor kappa B (NF-κB) signaling axis . This represents an IGF-independent function of IGFBP6.

  • Flow-dependent regulation: Disturbed flow (DF), a pro-atherogenic hemodynamic pattern, reduces IGFBP6 expression, potentially contributing to endothelial dysfunction and atherosclerosis initiation .

  • TNF interaction: Pro-inflammatory effects mediated by tumor necrosis factor (TNF) are reversed by IGFBP6 overexpression, suggesting IGFBP6 counteracts cytokine-induced inflammation .

  • Clinical relevance: IGFBP6 levels are significantly reduced in human atherosclerotic arteries and patient serum, indicating conserved mechanisms between mouse models and human disease .

This evidence establishes reduction of endothelial IGFBP6 as a predisposing factor in vascular inflammation and atherosclerosis, presenting a potential therapeutic target. The protective effect appears to be independent of its IGF binding capacity, highlighting the multifunctional nature of this protein.

What are the neurological and developmental phenotypes of IGFBP6 transgenic mice?

IGFBP6 transgenic mice exhibit several distinct neurological and developmental phenotypes:

Neurological phenotypes:

  • Reduced cerebellum size and weight: Transgenic mice expressing human IGFBP6 under the GFAP promoter showed approximately 25% reduction in cerebellum size and 35% reduction in weight .

  • Altered astrocyte differentiation: These mice had fewer differentiated, GFAP-expressing astrocytes compared to wild-type mice, suggesting IGFBP6 influences glial cell development .

  • Nuclear actions affecting differentiation: IGFBP6 can enter the nucleus and modulate cell differentiation and survival, potentially through interaction with transcription factors and DNA repair proteins .

Developmental phenotypes:

  • Growth retardation: Between birth and 1 month of age, transgenic mice had high levels of circulating human IGFBP6 and reduced plasma IGF-I levels, resulting in significantly reduced body weight .

  • Reproductive system effects: Transgenic females showed reduced litter size (27% reduction at 3 months of age; 66% reduction at 6 months), decreased ovary weight, fewer corpora lutea, and 50% reduction in circulating LH levels, suggesting altered ovulation and hypothalamic control .

These findings demonstrate that IGFBP6 influences both neural development and systemic growth parameters, consistent with its role in modulating both IGF-dependent and IGF-independent pathways during development.

How does IGFBP6 deficiency or overexpression affect cardiovascular pathophysiology in mice?

IGFBP6 plays a significant role in cardiovascular pathophysiology as demonstrated by both deficiency and overexpression studies:

Effects of IGFBP6 deficiency:

  • Enhanced atherosclerosis: IGFBP6-deficient mice exhibit aggravated diet-induced atherosclerosis .

  • Increased vulnerability to disturbed flow: These mice also show enhanced atherosclerotic lesion development in regions of disturbed flow (DF), which is a pro-atherogenic hemodynamic pattern .

  • Heightened endothelial inflammation: Reduction of IGFBP6 increases inflammatory molecule expression and monocyte adhesion to endothelial cells, key initiating events in atherosclerosis .

Effects of IGFBP6 overexpression:

  • Atheroprotection: Endothelial-cell-specific IGFBP6-overexpressing mice demonstrate protection against atherosclerosis .

  • Reduced vascular inflammation: IGFBP6 overexpression reverses pro-inflammatory effects mediated by disturbed flow and tumor necrosis factor (TNF) .

  • Anti-angiogenic effects: IGFBP6 overexpression inhibits angiogenesis in rhabdomyosarcoma xenografts and zebrafish embryos through IGF-independent mechanisms .

The molecular basis for these effects involves IGFBP6's interaction with the MVP–JNK/NF-κB signaling axis, which restrains inflammatory responses in endothelial cells . These findings establish IGFBP6 as a homeostasis-associated molecule in the cardiovascular system and suggest its potential as a therapeutic target for atherosclerosis.

What metabolic and growth phenotypes are associated with altered IGFBP6 expression in mice?

Alterations in IGFBP6 expression produce several distinct metabolic and growth phenotypes in mice:

Growth phenotypes:

  • Reduced body weight: Transgenic mice with high levels of circulating human IGFBP6 show significantly reduced body weight between birth and 1 month of age, associated with reduced plasma IGF-I levels .

  • Tissue-specific growth effects: Cerebellum size and weight are reduced by approximately 25% and 35%, respectively, in GFAP-promoter driven IGFBP6 transgenic mice .

Metabolic phenotypes:

  • IGF system modulation: IGFBP6 preferentially binds IGF-II over IGF-I, selectively inhibiting IGF-II actions including proliferation, survival, and differentiation across various cell types .

  • Altered astrocyte metabolism: Transgenic mice express fewer differentiated, GFAP-expressing astrocytes, which may impact brain energy metabolism given the role of astrocytes in providing metabolic support to neurons .

  • Reproductive metabolic effects: Female transgenic mice show altered reproductive physiology, including reduced ovary weight, fewer corpora lutea, and reduced circulating LH levels, suggesting impacts on the hypothalamic-pituitary-gonadal axis .

  • Potential senescence effects: Some studies suggest IGFBP6 may inhibit cellular senescence, although this finding contradicts other studies showing IGFBP6 decreases cell proliferation and survival .

These phenotypes demonstrate that IGFBP6 influences systemic and tissue-specific growth and metabolic parameters, with significant effects on developmental trajectories and adult physiology.

How can IGFBP6 mouse models inform therapeutic approaches for cardiovascular diseases?

IGFBP6 mouse models provide several insights that could inform therapeutic strategies for cardiovascular diseases:

  • Protective role identification: Studies showing that IGFBP6-deficient mice have aggravated atherosclerosis while endothelial-specific IGFBP6-overexpressing mice are protected establish IGFBP6 as a potential therapeutic target .

  • Mechanism elucidation: The identification of the MVP–JNK/NF-κB signaling axis as the pathway through which IGFBP6 executes its anti-inflammatory effects provides specific molecular targets for intervention .

  • Biomarker potential: The finding that IGFBP6 levels are significantly reduced in human atherosclerotic arteries and patient serum suggests IGFBP6 could serve as a biomarker for cardiovascular disease risk or progression .

  • Strategic approaches for therapy:

    • Recombinant IGFBP6 administration: Similar to recombinant mouse IGFBP6 protein used in experimental settings .

    • Gene therapy approaches: Targeted delivery of IGFBP6 to endothelial cells could provide localized protection.

    • Small molecule development: Compounds that stabilize endogenous IGFBP6 or mimic its anti-inflammatory actions.

    • Combination therapies: IGFBP6-based approaches could complement existing lipid-lowering or anti-inflammatory strategies.

  • Critical considerations: The dual IGF-dependent and IGF-independent functions of IGFBP6 require careful therapeutic design to target specific pathways without disrupting normal IGF signaling required for tissue homeostasis.

Mouse models provide a valuable platform for testing these therapeutic approaches before clinical translation, with the high degree of conservation between mouse and human IGFBP6 (70-85% sequence identity) supporting potential translational relevance .

What are the challenges in interpreting conflicting data from different IGFBP6 mouse models?

Researchers face several challenges when interpreting seemingly conflicting data from different IGFBP6 mouse models:

  • Promoter-specific expression patterns: Different transgenic models use distinct promoters (e.g., GFAP for CNS-specific expression), leading to varying tissue distribution of IGFBP6 that may produce phenotypic differences unrelated to IGFBP6's intrinsic function .

  • Developmental versus adult phenotypes: Constitutive overexpression or knockout models may induce developmental compensation that masks adult phenotypes, while inducible models reveal acute effects without compensatory mechanisms. This discrepancy requires careful experimental design and interpretation .

  • IGF-dependent versus IGF-independent effects: IGFBP6 has both IGF-dependent effects (through preferential binding to IGF-II) and IGF-independent actions (through various signaling pathways). Different experimental contexts may favor one set of functions over the other .

  • Cell type-specific responses: IGFBP6 effects vary significantly between cell types. For example:

    • Promotes migration in some tumor cells

    • Inhibits angiogenesis in endothelial cells

    • Suppresses differentiation in osteoblasts

    • Has context-dependent effects on apoptosis

  • Post-translational modifications: O-glycosylation and other modifications of IGFBP6 affect its stability and localization but not IGF binding. Different models may produce IGFBP6 with varying modifications, influencing results .

To address these challenges, researchers should:

  • Employ multiple complementary models (gain and loss of function)

  • Use tissue-specific and inducible systems

  • Directly compare IGF-dependent and independent functions

  • Analyze post-translational modifications

  • Validate findings across different experimental contexts

This comprehensive approach can help reconcile apparently conflicting data and build a more unified understanding of IGFBP6 biology.

What novel methodologies are emerging for studying IGFBP6 functions in mouse disease models?

Several cutting-edge methodologies are enhancing our ability to study IGFBP6 functions in mouse disease models:

  • CRISPR/Cas9 genome editing: Enables precise modification of the IGFBP6 gene to create:

    • Domain-specific mutations that selectively disrupt IGF binding versus IGF-independent functions

    • Conditional alleles for temporal and spatial control

    • Reporter knock-ins for real-time visualization of expression patterns

  • Single-cell transcriptomics: Provides unprecedented resolution of IGFBP6 expression patterns and responses across diverse cell populations within tissues. This approach can reveal cell type-specific effects that might be masked in bulk tissue analyses .

  • Spatial transcriptomics: Combines the high-throughput nature of RNA sequencing with spatial information, allowing visualization of IGFBP6 expression patterns in relation to tissue architecture and pathological features.

  • Intravital imaging: Using fluorescently tagged IGFBP6 or reporter systems to track its dynamics in living animals during disease progression, particularly valuable for studying its role in atherosclerosis or tumor development.

  • Proteomics approaches: Mass spectrometry-based techniques to identify IGFBP6 interacting partners across different tissues and disease states, helping elucidate tissue-specific mechanisms .

  • Organoid models: Derived from mouse tissues, organoids can serve as complex 3D models to study IGFBP6 functions in controlled yet physiologically relevant systems.

  • AAV-based delivery systems: Advanced viral vectors allow for rapid, tissue-specific modulation of IGFBP6 expression without the need for time-consuming generation of transgenic lines.

These emerging methodologies provide researchers with unprecedented tools to dissect the complex functions of IGFBP6 in normal physiology and disease states, potentially leading to new therapeutic applications.

How do findings from IGFBP6 mouse models translate to human cardiovascular disease?

Findings from IGFBP6 mouse models demonstrate significant translational relevance to human cardiovascular disease:

  • Conservation of expression patterns: The high sequence homology between mouse and human IGFBP6 (70-85%) suggests functional conservation . Recent studies confirm IGFBP6 is significantly reduced in human atherosclerotic arteries and patient serum, mirroring findings in mouse models .

  • Consistent atheroprotective mechanisms: In both mouse models and human endothelial cells, reduction of IGFBP6 increases inflammatory molecule expression and monocyte adhesion, while IGFBP6 overexpression reverses pro-inflammatory effects . This suggests conserved molecular mechanisms.

  • Signaling pathway conservation: The MVP–JNK/NF-κB signaling axis through which IGFBP6 executes anti-inflammatory effects appears to function similarly in human and mouse endothelial cells .

  • Clinical correlation data: Analysis of public datasets including GSE41571 (stable versus ruptured plaques), GSE163154 (intraplaque hemorrhage versus non-IPH), and others provides evidence that IGFBP6 expression is altered in human atherosclerotic disease, consistent with mouse model findings .

  • Potential biomarker application: The observed reduction of IGFBP6 in human patients with atherosclerosis suggests its potential use as a biomarker for cardiovascular disease risk or progression .

These correlations between mouse models and human disease underscore the potential value of IGFBP6-focused therapeutic strategies for cardiovascular diseases, although further clinical studies are needed to fully validate these approaches in humans.

How should researchers approach the design of experiments to validate mouse IGFBP6 findings in human systems?

To effectively validate mouse IGFBP6 findings in human systems, researchers should implement a comprehensive translational research strategy:

  • Cell type-specific comparative studies:

    • Compare effects of IGFBP6 modulation in matched mouse and human cell types (e.g., endothelial cells, astrocytes, reproductive tissues)

    • Use identical experimental conditions and readouts to facilitate direct comparison

    • Assess both IGF-dependent and IGF-independent functions across species

  • Functional domain analysis:

    • Identify conserved functional domains between mouse and human IGFBP6

    • Create chimeric proteins combining domains from both species to identify species-specific functions

    • Use site-directed mutagenesis to assess functional conservation of key residues

  • Clinical correlation studies:

    • Measure IGFBP6 levels in healthy individuals and patients with relevant pathologies (atherosclerosis, neurological disorders)

    • Correlate IGFBP6 levels with disease severity, progression, and outcomes

    • Analyze genetic variants in human IGFBP6 for association with disease phenotypes

  • Multi-omics integration:

    • Compare transcriptomic, proteomic, and metabolomic profiles between mouse models and human samples

    • Focus on pathways identified in mouse studies (e.g., MVP–JNK/NF-κB signaling) to assess conservation

    • Use pathway enrichment analysis to identify species-specific differences

  • Humanized mouse models:

    • Create mice expressing human IGFBP6 in place of endogenous mouse IGFBP6

    • Assess whether human IGFBP6 recapitulates or differs from effects of mouse IGFBP6

  • Validation in human tissues:

    • Use ex vivo human tissue explants to test effects of recombinant IGFBP6

    • Employ organoids derived from human tissues for functional studies

    • Analyze archived human specimens for IGFBP6 expression patterns in health and disease

This systematic approach enables robust translation of mouse findings to human systems while identifying potential species-specific differences that might affect therapeutic development.

What insights do IGFBP6 mouse studies provide for understanding neurological and reproductive disorders in humans?

IGFBP6 mouse studies provide valuable insights into potential mechanisms underlying human neurological and reproductive disorders:

Neurological disorders:

  • Developmental abnormalities: Transgenic mice expressing human IGFBP6 under the GFAP promoter showed reduced cerebellum size (25%) and weight (35%), suggesting IGFBP6 may contribute to neurodevelopmental disorders affecting cerebellar structure and function .

  • Astrocyte differentiation: These mice had fewer differentiated GFAP-expressing astrocytes, suggesting IGFBP6 influences glial development . Given the emerging role of astrocytes in neurodevelopmental and neurodegenerative diseases, this provides a potential mechanism for IGFBP6 involvement in conditions like autism spectrum disorders or Alzheimer's disease.

  • Nuclear actions affecting neuronal differentiation: IGFBP6 enters the nucleus where it can interact with transcription factors and modulate differentiation and survival . This suggests potential roles in disorders of neuronal development or neurodegeneration.

Reproductive disorders:

  • Ovulatory dysfunction: Female transgenic mice showed reduced litter size (27-66% reduction), marked decrease in ovary weight and number of corpora lutea, and 50% reduction in circulating LH levels . These findings suggest IGFBP6 may contribute to human disorders involving ovulatory dysfunction, including polycystic ovary syndrome or premature ovarian insufficiency.

  • Hypothalamic regulation: Reduced LH levels in transgenic mice suggest altered hypothalamic control, indicating IGFBP6 may influence the hypothalamic-pituitary-gonadal axis . This has implications for understanding neuroendocrine disorders affecting reproductive function.

  • Age-dependent fertility decline: The progressive reduction in litter size with age (27% at 3 months versus 66% at 6 months) in transgenic mice suggests IGFBP6 may contribute to age-dependent fertility decline in humans .

These insights provide mechanistic hypotheses that can guide investigation of IGFBP6's role in human neurological and reproductive disorders, potentially leading to novel diagnostic or therapeutic approaches.

How can researchers effectively analyze IGFBP6 expression data across different tissues and experimental conditions?

Effective analysis of IGFBP6 expression data requires a multi-faceted approach to account for tissue specificity, experimental conditions, and biological context:

  • Normalization strategies:

    • Use multiple reference genes for qPCR data normalization, selecting stable genes in the specific tissues being studied

    • Apply appropriate normalization methods for RNA-seq data (e.g., TPM, RPKM, or DESeq2 normalization)

    • Consider tissue-specific baseline expression levels when comparing across tissues

  • Statistical analysis considerations:

    • Account for potential batch effects and technical variation

    • Apply appropriate statistical tests based on data distribution

    • Use multiple testing correction for genome-wide or proteome-wide analyses

    • Consider power analysis to ensure sufficient sample size for detecting biologically meaningful differences

  • Integration of multi-omics data:

    • Correlate IGFBP6 mRNA with protein levels to assess post-transcriptional regulation

    • Examine post-translational modifications (O-glycosylation, phosphorylation, sulphation) that affect IGFBP6 function

    • Integrate with pathway analysis to understand context-specific regulation

  • Visualization techniques:

    • Use heatmaps for comparing expression across multiple tissues or conditions

    • Apply dimensionality reduction techniques (PCA, t-SNE) to identify patterns in high-dimensional data

    • Create tissue-specific expression atlases integrating spatial information

  • Validation across platforms:

    • Confirm key findings using complementary techniques (qPCR, Western blot, immunohistochemistry)

    • Compare with publicly available datasets (e.g., Mouse Genome Database)

    • Validate in independent biological replicates and different mouse strains

  • Temporal and spatial considerations:

    • Account for developmental stage-specific expression patterns

    • Consider circadian variation in expression

    • Analyze cell type-specific expression within heterogeneous tissues

This comprehensive approach enables robust interpretation of IGFBP6 expression data, facilitating meaningful comparisons across tissues and experimental conditions while minimizing confounding factors.

What are the key considerations for designing IGFBP6 functional assays in mouse models?

Designing effective functional assays for IGFBP6 in mouse models requires careful consideration of its dual IGF-dependent and IGF-independent activities:

  • Distinguishing IGF-dependent vs. IGF-independent effects:

    • Include appropriate controls with IGF-II or IGF-I addition/neutralization

    • Use IGFBP6 mutants with altered IGF binding affinity

    • Compare results in IGF-II knockout backgrounds versus wild-type mice

  • Context-specific functional readouts:

    • For proliferation/survival effects: BrdU incorporation, Ki67 staining, TUNEL assay, or cleaved caspase-3 detection in target tissues

    • For migration effects: Wound healing assays in cultured cells from transgenic mice, in vivo cell tracking

    • For anti-angiogenic effects: CD31 staining of vessel density, retinal angiogenesis assay, or matrigel plug assay

    • For anti-inflammatory effects: Adhesion molecule expression, monocyte adhesion, NF-κB activation assays

  • Tissue and cell type considerations:

    • Select appropriate cell isolation methods to maintain cellular phenotypes

    • Consider tissue-specific expression of IGFBP6 receptors and binding partners

    • Account for potential compensatory mechanisms in specific tissues

  • Temporal dynamics:

    • Design time-course experiments to capture both acute and chronic effects

    • Use inducible systems for temporal control of IGFBP6 expression

    • Consider developmental timing when interpreting phenotypes

  • Quantitative considerations:

    • Establish dose-response relationships for recombinant IGFBP6 administration

    • Use ELISA or similar methods to confirm transgene expression levels

    • Correlate phenotypic effects with IGFBP6 concentration

  • Pathway validation:

    • Include appropriate inhibitors of downstream pathways (e.g., MAP kinase inhibitors for migration studies, NF-κB inhibitors for inflammation studies)

    • Use phospho-specific antibodies to detect activation of key signaling nodes

    • Validate pathway components through genetic approaches (e.g., siRNA in cultured cells from transgenic mice)

These considerations help ensure that functional assays specifically address the biological roles of IGFBP6 in the appropriate cellular and physiological context.

How can researchers reconcile contradictory findings about IGFBP6 function in different experimental systems?

Reconciling contradictory findings about IGFBP6 function requires systematic analysis of experimental differences and biological context:

  • Systematic comparison of experimental conditions:

    • Create a detailed comparison table documenting key variables across studies:

      • Cell/tissue type

      • IGFBP6 concentration

      • Experimental duration

      • Presence of serum/growth factors

      • Glycosylation status of IGFBP6

      • Measurement techniques

  • Context-dependent mechanism analysis:

    • Investigate whether contradictory effects reflect truly different mechanisms in different contexts

    • For example, one study suggested IGFBP6 inhibits senescence while most other studies showed it decreases cell proliferation and survival . This may reflect cell type-specific responses.

  • Dose-dependent effects assessment:

    • Determine whether contradictions reflect different concentrations of IGFBP6

    • Create dose-response curves spanning concentrations used in different studies

    • Consider biphasic responses that might explain apparently contradictory results

  • Direct replication studies:

    • Perform side-by-side comparisons under identical conditions

    • Obtain biological materials from original researchers when possible

    • Document detailed protocols to identify subtle methodological differences

  • Integration with pathway knowledge:

    • Map contradictory findings to specific pathways (IGF-dependent vs. various IGF-independent mechanisms)

    • Determine whether contradictions reflect activation of different downstream pathways

    • Use pathway inhibitors to test specific mechanisms

  • Analysis of post-translational modifications:

    • Compare glycosylation, phosphorylation, or proteolytic processing status

    • O-glycosylation affects IGFBP6 stability and localization but not IGF binding

    • Proteolytic processing by different proteases may generate fragments with distinct activities

  • Meta-analysis approaches:

    • Apply formal meta-analysis techniques to quantitatively assess effect sizes across studies

    • Identify moderator variables that explain heterogeneity in results

    • Develop integrated models that incorporate context-dependent effects

This systematic approach can transform apparently contradictory findings into a more nuanced understanding of IGFBP6's context-dependent functions, ultimately advancing the field toward a unified model of IGFBP6 biology.

Product Science Overview

Structure and Function

IGFBP-6 is an O-linked glycoprotein that binds insulin-like growth factor-II (IGF-II) with a marked preferential affinity over IGF-I . This binding inhibits the activity of IGF-II, which is involved in various cellular processes such as proliferation, differentiation, and survival. IGFBP-6 has the strongest affinity for IGF-II among the IGFBPs, making it a critical regulator of IGF-II activity .

Biological Role

IGFBP-6 is synthesized in various tissues, including the central nervous system (CNS), and has been shown to inhibit proliferation in many cell types . It is involved in cell migration and the positive regulation of the stress-activated MAPK cascade . Additionally, IGFBP-6 has been implicated in obesity and other metabolic disorders .

Recombinant IGFBP-6

Recombinant human IGFBP-6 (rhIGFBP-6) has been synthesized using COS-7 monkey kidney cells that were transiently transfected with a eukaryotic expression vector containing a complementary DNA for IGFBP-6 . This recombinant protein is similar to IGFBP-6 purified from human cerebrospinal fluid in terms of IGF binding and O-glycosylation .

Research and Applications

Studies have shown that rhIGFBP-6 inhibits IGF-II-induced differentiation of L6A1 myoblasts in a dose-dependent manner . This inhibition is complete when rhIGFBP-6 is present in a slight molar excess. In contrast, rhIGFBP-6 does not affect IGF-I-induced differentiation, even when present in a five-fold molar excess . These findings highlight the preferential affinity of IGFBP-6 for IGF-II and its potential therapeutic applications in conditions where IGF-II activity needs to be modulated.

In the context of cancer research, IGFBP-6 has been shown to control the expansion of chemoresistant glioblastoma through paracrine IGF2/IGF-1R signaling . This suggests that IGFBP-6 could be a valuable target for developing new treatments for glioblastoma and other cancers.

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.