IGFBP2 Mouse

Insulin Like Growth Factor Binding Protein-2 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Overview of IGFBP2 Mouse

Insulin-like Growth Factor Binding Protein 2 (IGFBP2) Mouse refers to studies and tools involving the murine homolog of IGFBP2, a multifunctional protein implicated in development, metabolism, and oncogenesis. In mice, IGFBP2 is encoded by the Igfbp2 gene and shares ~82% amino acid homology with human IGFBP2 . Research on IGFBP2 Mouse encompasses:

  • Genetic models: Knockout (Igfbp2⁻/⁻) and transgenic mice .

  • Recombinant proteins: Tools for studying IGFBP2's structure and function .

  • Therapeutic peptides: Mimetics derived from IGFBP2 domains .

Cancer Development

IGFBP2 Mouse models reveal its oncogenic roles:

  • Glioma: Promotes tumor angiogenesis via RGD-integrin α5/β1 interactions .

  • Breast cancer: Enhances metastasis by recruiting endothelial cells via IGF1R activation .

  • Colorectal cancer: MMP-7 proteolysis releases IGF-II from IGFBP2, driving neoplastic transformation .

Bone Metabolism

  • Knockout mice: Male Igfbp2⁻/⁻ mice exhibit reduced bone volume/trabecular thickness (BV/TV: -35%) and impaired osteoblast differentiation .

  • Therapeutic peptide: PEG-HBD-1 (an HBD-derived peptide) rescues bone loss in ovariectomized mice by enhancing IGF-1/AKT signaling .

Neuroplasticity

  • JB2 peptide: An IGFBP2-mimetic peptide rescues synaptic deficits in Shank3 haploinsufficient mice, improving memory and motor function .

Key Research Findings from Mouse Models

Study FocusModelKey ResultsReference
Obesity and fatty liverIgfbp2⁻/⁻ miceReduced hepatic IGFBP2 expression (-40%) precedes obesity and insulin resistance .
Bone lossOvariectomized (OVX) miceOVX Igfbp2⁻/⁻ mice lose 25% more trabecular bone vs. wild-type .
Neurodevelopmental rescueShank3 mutant miceJB2 peptide normalizes synaptic plasticity and reduces seizure susceptibility .
Cancer signalingGlioma xenograftsIGFBP2 knockdown reduces tumor growth by 60% via integrin β1 inhibition .

Recombinant Mouse IGFBP2 Protein

  • Applications: ELISA, bioassays, and IGF-II binding studies (ED₅₀: 0.125–0.5 µg/mL) .

  • Structure: 271-amino acid mature protein (UniProt: P18065) .

Igfbp2 Knockout Mice

  • Phenotype: Sex-dependent effects; males show low bone mass, while females exhibit metabolic resilience post-ovariectomy .

  • Mechanism: Compensatory upregulation of IGFBP-1, -3, and -4 in knockout models .

Diagnostic and Therapeutic Peptides

  • JB2: Rescues ASD-related deficits via NMDA receptor and IGF2R activation .

  • PEG-HBD-1: Increases bone mineral density (+6.2%) in osteoporotic mice .

Product Specs

Description
Recombinant Mouse IGFBP2, produced in HEK293 cells, is a single glycosylated polypeptide chain encompassing amino acids 35-305. This protein, with a molecular mass of 30.3 kDa, comprises 277 amino acids. It features a 6-amino acid His tag at the C-terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
IGFBP2 protein solution at a concentration of 1 mg/ml. It is formulated in Phosphate-Buffered Saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), store the entire vial at 4°C. For extended periods, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing cycles should be avoided.
Purity
Purity exceeds 85.0% as determined by SDS-PAGE analysis.
Biological Activity
The ED50, determined by the ability to inhibit MCF-7 human breast cancer cell proliferation in the presence of Mouse IGF-2, is ≤ 0.7 µg/ml.
Synonyms

IBP-2, IGF-binding protein 2, IGFBP-2, mIGFBP-2, Igfbp-2, insulin-like growth factor binding protein 2 isoform 1, insulin-like growth factor binding protein 2.

Source

HEK293 cells.

Amino Acid Sequence

EVLFRCPPCT PERLAACGPP PDAPCAELVR EPGCGCCSVC ARQEGEACGV YIPRCAQTLR CYPNPGSELP LKALVTGAGT CEKRRVGTTP QQVADSDDDH SEGGLVENHV DGTMNMLGGG SSAGRKPLKS GMKELAVFRE KVNEQHRQMG KGAKHLSLEE PKKLRPPPAR TPCQQELDQV LERISTMRLP DDRGPLEHLY SLHIPNCDKH GRYNLKQCKM SLNGQRGECW CVNPNTGKPI QGAPTIRGDP ECHLFYNEQQ ETGGAHAQSV QHHHHHH.

Q&A

What is IGFBP2 and what are its primary functions in mice?

IGFBP2 is a member of the insulin-like growth factor binding protein family that modulates the biological effects of IGFs by controlling their distribution, function, and activity. In mice, IGFBP2 has both IGF-dependent inhibitory effects on normal somatic cell growth and IGF-independent activities that stimulate proliferation, survival, differentiation, and motility of various cell types .

The protein functions through multiple mechanisms:

  • Binding to cell surface integrin receptors, influencing cell mobility and proliferation

  • Interacting with Frizzled 8 and LDL receptor-related protein 6 to affect Wnt signaling

  • Supporting hematopoietic stem cell (HSC) maintenance and expansion

  • Regulating cell survival through pathways involving Bcl-2 and cell cycle inhibitors

Key experimental evidence demonstrates that IGFBP2 is essential for the HSC-supportive activity of activated endothelium and plays crucial roles in HSC survival and cycling in the bone marrow microenvironment .

How is IGFBP2 expressed in different mouse tissues and cell types?

IGFBP2 shows a distinctive expression pattern across different mouse tissues and cell types. Based on real-time RT-PCR analyses:

Cell PopulationRelative IGFBP2 Expression
HSCs (Lin⁻Sca-1⁺Kit⁺Flk2⁻CD34⁻)Baseline (1×)
BM CD45⁺ hematopoietic cells~2× higher than HSCs
CD45⁻ stromal cells~8× higher than HSCs
CD45⁻CD105⁺/CD44⁺/SSEA4⁺/CD29⁺/Sca-1⁺ cellsHigher expression than HSCs
CD45⁻CD31⁻ cellsHigher than CD45⁻CD31⁺ endothelial cells

IGFBP2 appears to be predominantly expressed by mesenchymal stromal cells rather than endothelial cells in the mouse bone marrow . Additionally, in both humans and mice, IGFBP2 is most highly expressed in astrocytes in the brain . This cell-specific expression pattern suggests important roles in the supportive microenvironments of various tissues.

What phenotypes are observed in IGFBP2-null mice?

IGFBP2-null mice exhibit several important hematopoietic phenotypes:

  • Decreased frequency of bone marrow Lin⁻Sca-1⁺Kit⁺ (LSK) cells

  • Reduced numbers of short-term HSCs/multipotent progenitors and long-term HSCs

  • Similar numbers of common myeloid progenitors (CMP), granulocyte-macrophage progenitors (GMP), megakaryocyte-erythroid progenitors (MEP), and common lymphoid progenitors (CLP) compared to wild-type mice

Beyond the hematopoietic system, IGFBP2-null mice display:

  • Lower spleen weights and reduced total splenic lymphocyte numbers

  • Decreased number and function of osteoblasts (in a gender-specific manner)

  • Normal total bone marrow cellularity

  • No overt developmental phenotype

The absence of major developmental defects despite specific cellular phenotypes suggests potential compensatory mechanisms during development.

What methods are most effective for measuring IGFBP2 protein levels in mouse samples?

Multiple validated approaches exist for detecting and quantifying IGFBP2 in mouse samples:

  • ELISA (Enzyme-Linked Immunosorbent Assay):

    • Solid-phase sandwich ELISA specifically designed for mouse IGFBP2 quantitation

    • Applicable for serum, plasma, and cell culture medium samples

    • Utilizes a matched antibody pair system with pre-coated microplates

    • Can exclusively recognize both natural and recombinant mouse IGFBP2

  • Western Blot Analysis:

    • Effective for semi-quantitative detection of IGFBP2 protein

    • Validated antibodies include goat anti-IGFBP2 (SC-6002; Santa Cruz Biotechnology)

    • Particularly useful for confirming IGFBP2 deficiency in knockout models

  • RT-PCR and Real-time RT-PCR:

    • For quantifying IGFBP2 expression at the mRNA level

    • Validated primer sets include QIAGEN cat no. QT00269542

    • Custom primers: 5′GGAGGGCGAAGCATGCGGCGTCTAC3′ and 5′GCCCATCTGCCGGTGCTGTTCATTGACCTT3′

When selecting a method, researchers should consider the specific research question, sample type, and whether protein or mRNA levels are more relevant to the experimental design.

How can researchers effectively compare IGFBP2 expression in different mouse models?

When comparing IGFBP2 expression across different mouse models, researchers should consider:

  • Tissue and cell type specificity:

    • IGFBP2 expression varies significantly between tissues and cell types

    • Cell sorting prior to analysis may be necessary for accurate comparisons

    • Single-cell approaches may reveal heterogeneity masked in bulk tissue analysis

  • Standardized sampling procedures:

    • Age and gender matching is critical (especially given gender-specific effects)

    • Consistent tissue collection protocols to minimize variability

    • Standardized processing times to avoid degradation

  • Multiple detection methods:

    • Combine mRNA and protein analyses to account for post-transcriptional regulation

    • Use in situ methods to preserve spatial information

    • Employ quantitative techniques like real-time RT-PCR rather than semi-quantitative methods

  • Statistical considerations:

    • Ensure adequate sample sizes based on preliminary data variance

    • Account for biological replicates versus technical replicates

    • Apply appropriate statistical tests for the data distribution

For example, studies comparing IGFBP2 expression in transgenic mouse models of tauopathy (P301L-tau) and Alzheimer's disease (TASTPM) with wild-type mice revealed significant differential expression in cortex but not hippocampus, highlighting the importance of region-specific analysis .

How does IGFBP2 regulate hematopoietic stem cell function in mice?

IGFBP2 regulates HSC function through several key mechanisms:

  • Survival regulation:

    • HSCs in IGFBP2-null mice show decreased survival

    • IGFBP2 influences the expression of anti-apoptotic factor Bcl-2

    • Loss of IGFBP2 leads to up-regulation of cell cycle inhibitors p21, p16, p19, p57, and PTEN

  • Cell cycle control:

    • IGFBP2 supports HSC cycling

    • IGFBP2-null mice show altered HSC cycling profiles

    • The balance between quiescence and proliferation is disrupted in the absence of IGFBP2

  • Environmental support:

    • IGFBP2 has minimal cell-autonomous effects on HSCs

    • Bone marrow stromal cells deficient for IGFBP2 have significantly decreased ability to support HSC expansion

    • IGFBP2 acts primarily through the microenvironment to influence HSC function

  • Domain-specific functions:

    • The C-terminus of IGFBP2 is essential for supporting HSC activity

    • The RGD domain appears less critical for HSC maintenance

    • IGFBP2's effects on HSCs are independent of IGF-IR-mediated signaling

These findings suggest that IGFBP2 primarily functions as an extrinsic factor in the bone marrow microenvironment to support HSC maintenance and expansion.

What are the experimental challenges in studying IGFBP2 effects on HSCs in mouse models?

Researchers face several significant challenges when investigating IGFBP2's role in HSC biology:

  • Distinguishing cell-autonomous versus non-cell-autonomous effects:

    • HSCs express minimal IGFBP2, while stromal cells produce higher levels

    • Requires careful experimental design using cell-specific knockouts or chimeric models

    • Competitive bone marrow transplantation assays with CD45 congenic markers are effective for assessing cell-intrinsic effects

  • Compensatory mechanisms:

    • Other IGFBPs may compensate for IGFBP2 deficiency

    • Changes in IGF bioavailability may mask direct IGFBP2 effects

    • Long-term versus acute loss of IGFBP2 may yield different phenotypes

  • Context-dependent functionality:

    • IGFBP2 has both IGF-dependent and IGF-independent functions

    • Different domains of IGFBP2 mediate different cellular effects

    • Experimental approaches must account for these distinct mechanisms

  • Technical limitations:

    • HSCs are rare and heterogeneous

    • Ex vivo studies may not recapitulate in vivo microenvironmental interactions

    • Long-term effects need to be distinguished from short-term responses

Effective approaches include using conditional knockout models, domain-specific mutants, and combined in vivo and ex vivo experimental systems to comprehensively assess IGFBP2 functions in HSC biology.

What is the relationship between IGFBP2 and Alzheimer's disease pathology in mouse models?

Research examining IGFBP2 in neurodegeneration reveals important connections with Alzheimer's disease (AD) pathology:

  • Differential expression in AD models:

    • Significant differential expression of IGFBP2 observed in transgenic mouse models of tauopathy (P301L-tau) and AD (TASTPM)

    • Expression changes are region-specific, occurring in cortex but not hippocampus

  • Correlation with tau pathology:

    • CSF IGFBP2 levels correlate with CSF tau levels in human studies

    • IGFBP2 levels associate with brain atrophy in non-hippocampal regions

    • This correlation suggests a potential mechanistic link between IGFBP2 and tau-related neurodegeneration

  • Cell-type specificity:

    • IGFBP2 is most highly expressed in astrocytes in both humans and mice

    • This suggests potential non-neuronal mechanisms in IGFBP2-related AD pathology

    • The astrocyte-specific expression may indicate roles in neuroinflammatory processes

  • Metabolic connections:

    • IGFBP2 represents a link between metabolic dysfunction and neurodegeneration

    • Brain insulin resistance and impaired insulin/IGF signaling are features of AD

    • IGFBP2 may be part of the dysregulated brain insulin/IGF signaling system in AD

These findings suggest that IGFBP2 could be an important mediator connecting metabolic dysfunction to neurodegenerative processes in AD, with potential relevance for therapeutic targeting.

How should researchers design experiments to investigate IGFBP2 contributions to neurodegeneration?

When designing experiments to study IGFBP2 in neurodegeneration, researchers should consider these methodological approaches:

  • Comprehensive phenotyping across multiple models:

    • Compare findings between different AD models (e.g., amyloid-based, tau-based)

    • Include age-matched controls to account for age-related changes in IGFBP2

    • Examine multiple brain regions separately due to region-specific effects

  • Cell type-specific analyses:

    • Given IGFBP2's high expression in astrocytes, cell-type-specific conditional knockout models are valuable

    • Single-cell or cell-sorted approaches to distinguish neuronal versus glial effects

    • Co-localization studies with cell-type markers and pathological features

  • Mechanistic pathway investigations:

    • Examine IGF-dependent versus IGF-independent mechanisms

    • Explore connections to insulin resistance pathways

    • Investigate relationships with both amyloid and tau pathology

  • Translational biomarker studies:

    • Correlate IGFBP2 levels with established AD biomarkers (Aβ, tau, p-tau)

    • Measure IGFBP2 in CSF, plasma, and brain tissue

    • Assess relationships with structural and functional neuroimaging

  • Intervention studies:

    • Manipulate IGFBP2 levels at different disease stages

    • Test domain-specific IGFBP2 fragments or inhibitors

    • Examine effects on both pathological features and behavioral outcomes

This multi-faceted approach will help establish whether IGFBP2 represents a causal factor, compensatory response, or biomarker in neurodegenerative processes.

How do post-translational modifications affect IGFBP2 function in mouse models?

Post-translational modifications (PTMs) of IGFBP2 represent an underexplored area with significant implications for function:

  • Proteolytic processing:

    • IGFBP2 can be cleaved by various proteases, generating fragments with distinct activities

    • The C-terminus of IGFBP2 has been shown to be essential for supporting HSC activity, independent of the RGD domain

    • Experimental approaches should include:

      • Western blot analysis with antibodies specific to different domains

      • Mass spectrometry to identify cleavage sites

      • Generation of cleavage-resistant IGFBP2 mutants

  • Phosphorylation:

    • Potential phosphorylation sites may regulate IGFBP2 binding to IGFs or cell surface receptors

    • Phosphorylation status could affect intracellular localization and function

    • Methodological considerations include:

      • Phospho-specific antibodies for detection

      • Phosphatase treatments to assess functional consequences

      • Site-directed mutagenesis of potential phosphorylation sites

  • Glycosylation:

    • N-linked and O-linked glycosylation may affect IGFBP2 stability and interactions

    • Glycosylation patterns could differ between tissues or disease states

    • Experimental approaches should include:

      • Glycosidase treatments

      • Lectin-based detection methods

      • Glycosylation site mutations

  • Oxidation/reduction:

    • IGFBP2 contains disulfide bonds that could be subject to redox regulation

    • Oxidative stress can trigger IGFBP2 uptake by cells

    • Research strategies should include:

      • Redox state analysis under different conditions

      • Mutation of cysteine residues

      • Assessment of IGFBP2 function under oxidative stress conditions

Understanding these PTMs will provide deeper insights into the context-specific functions of IGFBP2 in different tissues and disease states.

What are the molecular mechanisms underlying IGFBP2 overexpression in cancer models?

IGFBP2 overexpression is observed in multiple cancer types, and understanding the underlying mechanisms in mouse models can yield important insights:

  • Transcriptional regulation:

    • Investigate transcription factors that bind the IGFBP2 promoter in different cancer models

    • Examine whether oncogenic signaling pathways directly regulate IGFBP2 expression

    • Research approaches should include:

      • Promoter analysis using reporter assays

      • ChIP-seq to identify transcription factor binding

      • CRISPR-mediated disruption of regulatory elements

  • Epigenetic mechanisms:

    • IGFBP2 expression may be regulated by DNA methylation or histone modifications

    • These mechanisms could explain tissue-specific and context-dependent expression patterns

    • Experimental strategies should include:

      • Bisulfite sequencing of the IGFBP2 promoter

      • Histone modification ChIP at the IGFBP2 locus

      • Treatment with epigenetic modifiers to assess effects on expression

  • Signaling feedback loops:

    • IGFBP2 activates matrix metalloprotease 2, which contributes to cell invasiveness

    • This may create feed-forward mechanisms promoting continued IGFBP2 overexpression

    • Research approaches should include:

      • Pathway inhibition studies

      • Time-course analyses of IGFBP2 expression after pathway stimulation

      • In vivo models with inducible IGFBP2 expression

  • MicroRNA regulation:

    • Post-transcriptional regulation by miRNAs may control IGFBP2 levels

    • Different cancer types might exhibit distinct miRNA profiles affecting IGFBP2

    • Methodological considerations include:

      • miRNA target prediction and validation

      • miRNA modulation studies

      • Analysis of IGFBP2 mRNA stability

Understanding these mechanisms could identify potential therapeutic targets for cancers where IGFBP2 overexpression drives disease progression, with the correlation between IGFBP2 levels and tumor aggressiveness highlighting its clinical significance .

How can contradictory findings about IGFBP2 function in different mouse models be reconciled?

Researchers encountering contradictory data about IGFBP2 should consider these methodological approaches to reconciliation:

  • Context-dependent functionality:

    • IGFBP2 has both IGF-dependent inhibitory effects and IGF-independent stimulatory effects

    • Different tissues may exhibit opposite responses to IGFBP2

    • Reconciliation strategies should include:

      • Side-by-side comparison of models using identical methodologies

      • Analysis of cellular context and microenvironment differences

      • Domain-specific mutants to isolate particular functions

  • Developmental timing effects:

    • IGFBP2's roles may differ during development versus adult homeostasis

    • Compensatory mechanisms may mask phenotypes in constitutive knockout models

    • Research approaches should include:

      • Inducible/conditional models to control timing of IGFBP2 deletion

      • Developmental time course studies

      • Acute versus chronic loss-of-function comparisons

  • Strain-specific genetic modifiers:

    • Background strain differences can significantly impact phenotypes

    • IGFBP2 effects may be influenced by strain-specific alleles of interacting genes

    • Experimental strategies should include:

      • Backcrossing to multiple pure genetic backgrounds

      • Genetic mapping of modifier loci

      • Analysis of IGFBP2 function in diverse genetic contexts

  • Technical considerations:

    • Different antibodies may recognize distinct forms or epitopes of IGFBP2

    • Assay conditions can affect IGF binding and other protein interactions

    • Methodological approaches should include:

      • Validation with multiple detection methods

      • Recombinant protein controls

      • Careful documentation of experimental conditions

Systematic application of these approaches can help distinguish true biological complexity from technical artifacts, providing a more nuanced understanding of IGFBP2's multifaceted functions.

What are the most promising research directions for IGFBP2 in mouse models?

Based on current knowledge gaps and potential applications, several research directions merit prioritization:

  • Cell type-specific conditional models:

    • Generate and characterize astrocyte-specific IGFBP2 knockout models for neurodegeneration studies

    • Develop stromal cell-specific deletion models to further elucidate non-cell-autonomous effects on HSCs

    • Create inducible overexpression models to assess therapeutic potential

  • Domain-specific functional analysis:

    • Further investigate the C-terminal domain that supports HSC activity

    • Determine structural requirements for astrocyte-specific functions in neurodegeneration

    • Develop domain-specific blocking antibodies or peptides for targeted interventions

  • Translational biomarker development:

    • Validate IGFBP2 as a biomarker for early detection or progression monitoring in neurodegeneration

    • Examine IGFBP2 correlations with therapeutic responses in disease models

    • Establish standardized assays for IGFBP2 quantification across biofluids

  • Therapeutic modulation strategies:

    • Test IGFBP2 supplementation for HSC transplantation enhancement

    • Evaluate IGFBP2 inhibition in cancer models

    • Explore cell-specific targeting approaches to minimize off-target effects

These directions will advance both fundamental understanding of IGFBP2 biology and potential clinical applications, building on the established roles in stem cell maintenance, cancer, and neurodegeneration.

What methodological innovations would advance IGFBP2 research in mouse models?

Several methodological innovations could significantly advance IGFBP2 research:

  • In vivo imaging approaches:

    • Develop fluorescent or bioluminescent IGFBP2 reporter mice

    • Apply intravital microscopy to observe IGFBP2 dynamics in living tissues

    • Create biosensors to detect IGFBP2-protein interactions in real-time

  • Single-cell multi-omics:

    • Apply single-cell RNA-seq to identify cell populations responding to IGFBP2

    • Use spatial transcriptomics to map IGFBP2 expression in tissue microenvironments

    • Integrate proteomics and transcriptomics to capture post-transcriptional regulation

  • CRISPR-based functional screening:

    • Apply CRISPR activation/interference to identify regulators of IGFBP2

    • Screen for genes that modify IGFBP2-dependent phenotypes

    • Create precise point mutations to map functional domains

  • Humanized mouse models:

    • Generate mice expressing human IGFBP2 to better model human disease

    • Create patient-derived xenograft models to study IGFBP2 in human cancer

    • Develop models that recapitulate human-specific regulatory mechanisms

Product Science Overview

Introduction

Insulin-Like Growth Factor Binding Protein-2 (IGFBP-2) is a member of the insulin-like growth factor binding protein family, which plays a crucial role in modulating the activity of insulin-like growth factors (IGFs). IGFBP-2 is known for its high affinity for IGF-I and IGF-II, which are essential for cell growth, development, and metabolism. The recombinant form of IGFBP-2 derived from mice is widely used in research to study its biological functions and potential therapeutic applications.

Structure and Function

IGFBP-2 is a glycoprotein consisting of approximately 328 amino acids. It contains three distinct domains: the N-terminal domain, the central domain, and the C-terminal domain. Each domain contributes to the protein’s ability to bind IGFs and modulate their activity. The N-terminal domain is responsible for high-affinity binding to IGFs, while the central domain contains a nuclear localization signal that allows IGFBP-2 to enter the nucleus and influence gene expression. The C-terminal domain is involved in interactions with cell surface receptors and extracellular matrix components.

Biological Role

IGFBP-2 plays a multifaceted role in regulating IGF activity. By binding to IGFs, IGFBP-2 can inhibit their interaction with IGF receptors, thereby modulating their mitogenic and metabolic effects. Additionally, IGFBP-2 can enhance IGF activity by protecting IGFs from degradation and extending their half-life in circulation. Beyond its role in IGF regulation, IGFBP-2 has been implicated in various cellular processes, including cell migration, invasion, and angiogenesis. It is also involved in the regulation of glucose metabolism and has been linked to metabolic disorders such as diabetes.

Preparation Methods

The recombinant form of IGFBP-2 (Mouse) is typically produced using recombinant DNA technology. The gene encoding IGFBP-2 is cloned into an expression vector, which is then introduced into a suitable host cell line, such as Escherichia coli or Chinese hamster ovary (CHO) cells. The host cells are cultured under optimal conditions to express the recombinant protein, which is subsequently purified using techniques such as affinity chromatography and gel filtration. The purified recombinant IGFBP-2 is then characterized to ensure its structural and functional integrity.

Applications in Research

Recombinant IGFBP-2 (Mouse) is widely used in research to investigate its biological functions and potential therapeutic applications. It is employed in studies exploring the role of IGFBP-2 in cancer, where it has been shown to influence tumor growth, metastasis, and response to therapy. Additionally, IGFBP-2 is used in research on metabolic disorders, such as diabetes and obesity, to understand its impact on glucose metabolism and insulin sensitivity. The protein is also utilized in studies examining its role in cardiovascular diseases, neurodegenerative disorders, and tissue regeneration.

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.