LR3 IGF1 Human

LR3 Insulin Like Growth Factor-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Mechanism of Action

LR3 IGF-1 Human binds to the IGF-1 receptor and insulin receptor with affinity comparable to native IGF-1 but exhibits lower affinity for IGF-binding proteins (IGFBPs) . This reduced binding to IGFBPs prolongs its bioavailability and enhances its anabolic effects.

Key Pathways and Effects:

  1. Protein Synthesis:

    • Activates the PI3K/Akt/mTOR pathway, stimulating ribosomal protein synthesis in muscle and other tissues .

    • Inhibits myostatin, a negative regulator of muscle growth .

  2. Muscle Cell Dynamics:

    • Hypertrophy: Increases cross-sectional area of existing muscle fibers.

    • Hyperplasia: Promotes proliferation and differentiation of satellite cells, enabling new muscle fiber formation .

  3. Metabolic Regulation:

    • Reduces glucose uptake in adipose tissue, redirecting energy substrates to muscle .

    • Lowers insulin levels while maintaining glucose homeostasis .

Muscle Growth and Regeneration

Studies in animal models and human trials demonstrate:

  • Adults: A 6-month trial in older males showed an 8.8% increase in lean muscle mass and 14.4% reduction in adipose tissue with IGF-1 therapy, though LR3 is 2–3× more potent .

  • Fetal Development: In sheep models, LR3 IGF-1 infusion increased heart, spleen, and adrenal gland weights without altering fetal body weight, suggesting organ-specific growth .

Model/StudyDurationKey OutcomesSource
Aging human males6 months↑8.8% lean mass, ↓14.4% fat, ↑1.6% bone density
Fetal sheep1 week↑ Heart, spleen, adrenal weights; ↓ insulin
Cultured myoblastsIn vitro↑ Protein synthesis, ↓ protein breakdown

Therapeutic Potential

LR3 IGF-1 Human is investigated for:

  • Muscle-wasting diseases: HIV-associated cachexia, sarcopenia.

  • Growth disorders: Turner syndrome, pituitary tumors.

  • Organ repair: Cardiac hypertrophy, liver regeneration .

Product Specs

Introduction
Insulin-like growth factor-1 (IGF-1) is a key hormone that regulates growth. Growth hormone (GH) normally stimulates the production and release of IGF-1 from the liver and other tissues. IGF-1 then activates the Type 1 IGF receptor, similar to the insulin receptor, in target tissues, initiating intracellular signaling pathways that promote growth. IGF-1 also plays a role in metabolism by enhancing the uptake of nutrients like glucose, fatty acids, and amino acids to support tissue growth.
Description
LR3 is a long-acting analog of human IGF-1 engineered for mammalian cell culture, supporting large-scale biopharmaceutical production. This recombinant human LR3 Insulin Like Growth Factor-1, produced in E. coli, is a single, non-glycosylated polypeptide chain with 83 amino acids and a molecular weight of 9.1 kDa.
Physical Appearance
White, sterile-filtered powder obtained by lyophilization.
Formulation

The product is lyophilized from a 0.2µm filtered solution concentrated in 20mM PB, at a pH of 7.2.

Solubility
To reconstitute lyophilized LR3 IGF1, it is recommended to dissolve it in sterile 18M-cm H2O at a concentration of 100µg/ml. This solution can be further diluted with other aqueous solutions as needed.
Stability
While lyophilized LR3 IGF1 remains stable at room temperature for up to 3 weeks, it is best stored desiccated below -18°C. Reconstituted LR3 IGF1 should be kept at 4°C for 2-7 days or stored below -18°C for extended periods. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity

The purity is determined to be greater than 98.0% using SDS-PAGE analysis.

Biological Activity
The ED50, determined by measuring the stimulation of protein synthesis in L6 myoblasts, is less than 10ng/ml. This corresponds to a specific activity of 100,000 units/mg.
Synonyms
R3 IGF1, R3 IGF-1, R3IGF1, R3IGF-1, LONG IGF1, LONG IGF-1, LONG R3 IGF1, LONG R3IGF1, LONG R3 IGF-1, LONG R3IGF-1.
Source
Escherichia Coli.
Amino Acid Sequence
MFPAMPLSSLFVNGPRTLCGAELVDALQFVCGDRGFYFNKPTGYGSSSRRAPQTGIV DECCFRSCDLRRLEMYCAPLKPAKSA.

Q&A

What is LR3 IGF-I and how does it differ from native human IGF-I?

LR3 IGF-I is an 83 amino acid synthetic analog of human IGF-I, specifically engineered for enhanced biological activity in research applications. It differs from native IGF-I through two key modifications: (1) substitution of an arginine for glutamine at position 3 of the mature IGF-I sequence, and (2) addition of a 13 amino acid N-terminal extension peptide derived from methionyl porcine Growth Hormone . These structural changes significantly reduce its binding affinity to IGF binding proteins (IGFBPs) while maintaining similar binding affinity to IGF-I and insulin receptors . Consequently, LR3 IGF-I exhibits an extended half-life of approximately 20-30 hours compared to native IGF-I's 12-15 minutes in circulation, resulting in substantially increased biological potency both in vitro and in vivo . This enhanced stability makes it particularly valuable for research applications requiring sustained IGF-I receptor activation.

What is the molecular structure and physical properties of LR3 IGF-I?

LR3 IGF-I is a 9.2 kDa protein comprising the complete human IGF-I sequence (Gly49-Ala118) with the Glu51Arg substitution, plus the 13-amino acid N-terminal extension . Mass spectrometry analysis through ESI confirms a molecular mass of approximately 9112 Da, which closely aligns with the calculated theoretical mass of 9118 Da . The protein appears as a single band at approximately 7-8 kDa when analyzed by SDS-PAGE under reducing conditions . The amino acid structure can be represented as:

N-terminus (Extension)Human LR3 IGF-I (Gly49-Ala118 with Glu51Arg substitution)
MFPAMPLSSLFVNModified human IGF-I sequence with Arg at position 3

The protein is typically supplied in lyophilized form from a phosphate buffer solution (pH 7.2) and requires reconstitution in sterile PBS at concentrations of approximately 500 μg/mL for experimental use .

What are the primary research applications for LR3 IGF-I?

LR3 IGF-I was specifically developed for supplementation of mammalian cell culture systems to enhance cell survival and proliferation . Its primary research applications include:

  • Cell culture optimization: Serves as an alternative to insulin for promoting cell growth and protein production in serum-free or serum-reduced conditions

  • Signal transduction studies: Used to investigate IGF-I receptor-mediated signaling pathways with reduced interference from IGFBPs

  • Cellular growth and differentiation research: Facilitates studies on mechanisms of protein synthesis, cell hypertrophy, and metabolic regulation

  • Apoptosis inhibition: Enables research on cell survival pathways and programmed cell death prevention

  • Nutrient uptake analysis: Supports investigations of glucose and amino acid transport mechanisms

The ED50 for cell proliferation effects in models such as the MCF-7 human breast cancer cell line is typically 0.3-1.5 ng/mL, demonstrating its high potency in experimental systems .

What are the optimal protocols for using LR3 IGF-I in cell culture systems?

For optimal implementation of LR3 IGF-I in cell culture systems, researchers should consider the following methodological approach:

Concentration optimization: LR3 IGF-I is significantly more potent than insulin and native IGF-I, requiring careful titration. Typical effective concentrations range from 0.3-50 ng/mL depending on cell type and experimental goals . Start with lower concentrations (1-5 ng/mL) and determine the minimum effective dose for your specific cell line through dose-response experiments.

Medium formulation: LR3 IGF-I can be used in both serum-containing and serum-free media. For serum-free applications, supplement with appropriate proteins (albumin, transferrin) to stabilize the peptide . When replacing insulin in culture media, typical LR3 IGF-I concentrations are 50-100 times lower than insulin concentrations while achieving equivalent or superior results .

Reconstitution and storage: Reconstitute lyophilized LR3 IGF-I in sterile PBS at 500 μg/mL and store aliquots at -20°C or -80°C to avoid repeated freeze-thaw cycles . Working solutions should be prepared fresh in complete culture medium. Stability at 37°C typically extends 3-5 days in cell culture conditions.

Administration schedule: For long-term cultures, replenish LR3 IGF-I every 48-72 hours to maintain effective concentrations, as degradation may occur despite the extended half-life compared to native IGF-I.

How can researchers quantify LR3 IGF-I in experimental samples?

Accurate quantification of LR3 IGF-I in experimental samples is essential for standardization and reproducibility. Current validated methodologies include:

ELISA-based quantification: Specialized RapidTM ELISA kits have been developed specifically for LR3 IGF-I quantification, utilizing mouse monoclonal anti-LR3 IGF-I capture antibodies and biotinylated detection antibodies . These assays provide sensitive and specific quantification in less than 3 hours with detection limits in the low ng/mL range. The complete workflow involves:

  • Sample preparation (typically dilution in appropriate buffers)

  • Binding to pre-coated capture antibodies

  • Detection with biotinylated detection antibodies

  • Signal amplification using HRP-conjugated streptavidin

  • Colorimetric detection with TMB substrate

  • Quantification against a standard curve of known LR3 IGF-I concentrations

Mass spectrometry: For advanced research requiring higher specificity, ESI-MS can be employed to detect LR3 IGF-I with molecular weight confirmation at approximately 9112 Da . This approach is particularly valuable when analyzing complex biological matrices or when assessing structural integrity.

Bioactivity assays: Functional quantification using proliferation assays in responsive cell lines such as MCF-7 breast cancer cells, where the ED50 for LR3 IGF-I typically falls between 0.3-1.5 ng/mL . This approach provides information on both concentration and biological activity.

What signaling pathways are activated by LR3 IGF-I and how do they compare to native IGF-I?

LR3 IGF-I activates several intracellular signaling cascades through binding to the IGF-1 receptor, with some notable differences from native IGF-I due to its reduced IGFBP interaction:

Primary signaling pathways activated:

  • PI3K-Akt pathway: LR3 IGF-I potently activates the phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) pathway, which is critical for protein synthesis, glucose metabolism, and anti-apoptotic effects . The sustained activation of this pathway is typically more pronounced with LR3 IGF-I compared to native IGF-I due to reduced sequestration by IGFBPs.

  • MAPK/ERK signaling: Mitogen-activated protein kinase cascades are stimulated, promoting cell proliferation and differentiation . The kinetics of ERK1/2 phosphorylation may differ between LR3 IGF-I and native IGF-I, with LR3 IGF-I potentially inducing more sustained activation.

  • mTOR pathway: LR3 IGF-I activates the mammalian target of rapamycin complex, regulating protein synthesis and cellular growth processes . This pathway is particularly relevant in research on muscle hypertrophy and cellular energy metabolism.

Comparative signaling dynamics:

ParameterLR3 IGF-INative IGF-I
Receptor binding affinitySimilar to IGF-IBaseline
IGFBP interactionSignificantly reducedHigh affinity
Signal durationExtended (hours)Shorter (minutes)
Pathway activation thresholdLower concentrations effectiveHigher concentrations required
Cell type specificitySimilar receptor distributionSimilar receptor distribution

The reduced IGFBP binding of LR3 IGF-I results in more direct receptor activation and more prolonged signaling, making it particularly valuable for studying the intrinsic effects of IGF-1 receptor activation without the confounding effects of binding protein interactions .

What are the key controls and variables to consider when designing experiments with LR3 IGF-I?

When designing robust experiments involving LR3 IGF-I, researchers should implement the following controls and account for these experimental variables:

Essential controls:

  • Negative controls: Include untreated cells to establish baseline cellular activities and responses.

  • Native IGF-I control: Compare effects with equimolar concentrations of native IGF-I to quantify the enhanced potency of LR3 IGF-I.

  • Insulin control: When replacing insulin in media formulations, include insulin-supplemented conditions at standard concentrations for comparison .

  • Receptor blocking: Include conditions with IGF-1 receptor antagonists or neutralizing antibodies to confirm specificity of observed effects.

  • Signaling pathway inhibitors: Use specific inhibitors (e.g., wortmannin for PI3K, U0126 for MEK/ERK) to dissect the contribution of individual downstream pathways.

Critical experimental variables:

  • Cell density and passage number: Receptor expression and response magnitude can vary significantly with cell confluency and passage history.

  • Serum components: Even trace amounts of serum can contribute IGFBPs that potentially interact with LR3 IGF-I, albeit at reduced levels compared to native IGF-I.

  • Exposure duration: Due to its extended half-life, time-course experiments should extend beyond timepoints typically used for native IGF-I .

  • Concentration range: The high potency of LR3 IGF-I necessitates careful titration, as concentrations effective for one cellular response may differ from those optimal for another.

  • Cell type specificity: Different cell types express varying levels of IGF-1 receptor and insulin receptor, potentially altering response thresholds.

How should researchers approach contradictory results when comparing LR3 IGF-I to native IGF-I?

When encountering contradictory experimental outcomes between LR3 IGF-I and native IGF-I, researchers should systematically investigate the following potential sources of discrepancy:

Binding protein interference: The reduced IGFBP binding of LR3 IGF-I represents its primary functional difference from native IGF-I . Quantify IGFBP levels in your experimental system using appropriate ELISAs or Western blots. Contradictory results may stem from varying IGFBP expression across cell types or experimental conditions.

Receptor expression profiling: Determine the relative expression levels of IGF-1 receptor (IGF1R) and insulin receptor (IR) in your model system, as LR3 IGF-I and native IGF-I may differ in their relative affinities for these receptors and receptor hybrids. RNA sequencing or receptor-specific antibodies can quantify these differences.

Signaling duration analysis: Investigate temporal dynamics through time-course experiments examining phosphorylation status of key signaling molecules (e.g., Akt, ERK1/2, S6K) at multiple timepoints ranging from minutes to hours . The extended half-life of LR3 IGF-I may result in differences observable only at later timepoints.

Dose-response relationships: Perform parallel dose-response analyses for both molecules across a wide concentration range (0.1-100 ng/mL). Contradictory findings may result from comparing non-equivalent portions of their respective dose-response curves.

Experimental system context: Consider the presence of confounding factors such as autocrine/paracrine growth factor production, which may mask or potentiate differences between LR3 IGF-I and native IGF-I.

What are the appropriate storage and handling protocols to maintain LR3 IGF-I stability?

Maintaining the structural integrity and biological activity of LR3 IGF-I requires careful attention to storage and handling procedures:

Reconstitution guidelines:

  • Lyophilized LR3 IGF-I should be stored at -20°C upon receipt.

  • Reconstitute to 500 μg/mL in sterile, filtered PBS (pH 7.2-7.4) .

  • Allow the protein to dissolve completely through gentle swirling rather than vortexing, which may damage the protein structure.

  • For enhanced stability, consider adding a carrier protein (0.1-1% BSA) for dilute solutions.

Storage recommendations:

  • Prepare single-use aliquots immediately after reconstitution to avoid repeated freeze-thaw cycles.

  • Store reconstituted aliquots at -20°C for short-term (1-2 months) or -80°C for long-term storage (6-12 months) .

  • Record lot numbers, reconstitution dates, and freeze-thaw events for each aliquot to maintain experimental consistency.

Stability considerations:

  • Avoid more than 2-3 freeze-thaw cycles, as each cycle can result in approximately 10-15% activity loss.

  • When thawing, allow the protein to warm to room temperature gradually; rapid temperature changes can affect tertiary structure.

  • In cell culture media at 37°C, LR3 IGF-I typically maintains activity for 3-5 days, significantly longer than native IGF-I.

Quality control measures:

  • Periodically verify protein activity through bioassays using established responsive cell lines (e.g., MCF-7) .

  • For critical applications, confirm protein integrity through SDS-PAGE or mass spectrometry prior to use in key experiments.

How does LR3 IGF-I compare to insulin for supporting protein production in mammalian cell culture?

LR3 IGF-I offers several advantages over insulin for supporting recombinant protein production in mammalian expression systems, with important methodological differences:

Comparative efficacy:

LR3 IGF-I typically achieves equivalent or superior productivity to insulin at concentrations 50-100 times lower . This differential is attributed to:

  • Greater specificity for the IGF-1 receptor, which more directly activates anabolic and anti-apoptotic pathways

  • Extended half-life in culture media, requiring less frequent supplementation

  • Reduced interaction with binding proteins, allowing more direct cellular effects

Implementation strategies:

ParameterLR3 IGF-IInsulin
Typical effective concentration5-20 ng/mL5-10 μg/mL
Supplementation frequencyEvery 2-3 daysEvery 1-2 days
Receptor activation profilePrimarily IGF1R > IRIR > IGF1R
Downstream pathway biasMore balanced PI3K/MAPKMore IR-specific pathways
Cost per unit biological effectInitially higher but potentially more economical long-termLower unit cost but higher total usage

Cell line-specific considerations:

The relative advantage of LR3 IGF-I over insulin varies by cell line and depends on the relative expression of IGF1R versus IR. CHO cells typically show pronounced benefits from LR3 IGF-I supplementation due to their IGF1R expression profile . Hybridoma and HEK293 cells also demonstrate enhanced viability and productivity with LR3 IGF-I compared to insulin at equivalent signal-generating concentrations.

Production output impact:

LR3 IGF-I supplementation has been reported to increase recombinant protein yields by 15-40% compared to insulin in various expression systems, primarily through extended culture viability and reduced apoptosis rather than increased per-cell productivity .

What methodologies are recommended for studying LR3 IGF-I effects on cellular metabolism?

To comprehensively investigate LR3 IGF-I-mediated effects on cellular metabolism, researchers should employ these methodological approaches:

Glucose metabolism assessment:

  • Glucose uptake analysis: Measure uptake of radiolabeled 2-deoxyglucose or fluorescent glucose analogs (2-NBDG) following LR3 IGF-I treatment at various timepoints (30 min - 24h).

  • Glycolytic flux measurement: Employ extracellular acidification rate (ECAR) analysis using platforms such as Seahorse XF to quantify changes in glycolytic activity in real-time following LR3 IGF-I exposure .

  • Glucose transporter translocation: Track GLUT4 trafficking in relevant cell types using fluorescently-tagged constructs or cell surface biotinylation assays to determine if LR3 IGF-I enhances glucose transporter recruitment.

Amino acid metabolism investigation:

  • Amino acid uptake: Measure uptake of radiolabeled or fluorescently labeled amino acids (particularly branched-chain amino acids) following LR3 IGF-I treatment .

  • Protein synthesis quantification: Use puromycin incorporation (SUnSET method) or heavy isotope labeling combined with mass spectrometry to measure de novo protein synthesis rates.

  • Amino acid transporter expression: Quantify changes in expression of key transporters (e.g., LAT1, SNAT2) using qPCR and Western blotting.

Metabolomic profiling:

Conduct untargeted LC-MS/MS metabolomic analysis of cells treated with LR3 IGF-I versus controls to identify broader metabolic pathway alterations beyond the canonical glucose and amino acid effects .

What are the methodological considerations for studying LR3 IGF-I effects across different cell types?

Investigation of LR3 IGF-I effects across diverse cell types requires careful methodological planning to account for cell-specific differences in receptor expression, signaling dynamics, and functional outcomes:

Receptor profiling strategy:

  • Quantify IGF1R and IR expression levels across your cell panel using qPCR and flow cytometry or Western blotting.

  • Determine receptor hybrid (IGF1R/IR) formation through co-immunoprecipitation studies, as these hybrids may respond differently to LR3 IGF-I compared to homodimeric receptors.

  • Measure IGFBP expression and secretion profiles, as some cell types may produce binding proteins that modulate LR3 IGF-I availability despite its reduced IGFBP affinity.

Cell type-specific dose optimization:

Cell TypeRecommended Starting Concentration RangePrimary Response Parameters
Myoblasts/Myotubes5-50 ng/mLDifferentiation, protein synthesis, glucose uptake
Fibroblasts1-20 ng/mLProliferation, collagen production, migration
Epithelial cells2-30 ng/mLProliferation, barrier function, polarization
Neuronal cells10-100 ng/mLNeurite outgrowth, survival, synaptic plasticity
Hepatocytes5-50 ng/mLMetabolic enzyme regulation, protein production

Response heterogeneity analysis:

  • Temporal dynamics: Different cell types may exhibit varying temporal response patterns to LR3 IGF-I. Design time-course experiments spanning 5 minutes to 72 hours to capture both rapid signaling events and delayed transcriptional responses.

  • Pathway dominance: Systematically inhibit key pathways (PI3K/Akt, MEK/ERK, mTOR) to determine which predominates in mediating LR3 IGF-I effects in each cell type. Use pathway-specific inhibitors at established IC50 concentrations.

  • Functional readouts: Select cell type-appropriate functional assays relevant to the physiological role of IGF signaling in that cell type (e.g., protein synthesis for muscle cells, glucose production for hepatocytes, branching morphogenesis for epithelial cells).

  • Single-cell approaches: For heterogeneous populations, consider single-cell RNA-seq or CyTOF analysis to identify differential responsiveness within subpopulations that might be masked in bulk analyses.

What are the current limitations in LR3 IGF-I research methodologies?

Current methodological limitations in LR3 IGF-I research include challenges in standardization across laboratories, incomplete understanding of its interactions with the cellular microenvironment, and technical constraints in visualizing real-time receptor dynamics. These limitations highlight opportunities for methodological innovation, particularly in developing more sophisticated reporter systems, improved in vivo tracking methods, and standardized protocols for quantitative comparison across research groups. Addressing these limitations will be essential for translating mechanistic findings into practical applications for cell culture optimization and biomedical research.

Product Science Overview

Structure and Modifications

LR3 IGF-1 differs from native IGF-1 in two significant ways:

  1. Amino Acid Substitution: The third amino acid in the sequence of IGF-1 is replaced with an arginine residue instead of the naturally occurring glutamic acid. This substitution is referred to as “arginine 3”.
  2. Extended N-Terminus: LR3 IGF-1 has an additional 13 amino acids at its N-terminus, resulting in a total of 83 amino acids compared to the 70 amino acids in native IGF-1 .

These modifications result in LR3 IGF-1 having a significantly reduced affinity for insulin-like growth factor-binding proteins (IGFBPs), which normally inhibit the biological activity of IGF-1. Consequently, LR3 IGF-1 exhibits enhanced metabolic stability and potency, making it approximately three times more potent than native IGF-1 .

Biological Activity and Applications

LR3 IGF-1 retains the pharmacological activity of IGF-1 as an agonist of the IGF-1 receptor. This receptor is involved in various cellular processes, including cell growth, differentiation, and survival. The enhanced stability and potency of LR3 IGF-1 make it particularly useful in research settings where prolonged activity is required .

In addition to its research applications, LR3 IGF-1 is also used in the production of recombinant biopharmaceuticals. Its ability to support mammalian cell culture makes it an essential component in the large-scale manufacturing of these products .

Preparation and Production

LR3 IGF-1 is typically produced using recombinant DNA technology. The gene encoding the modified IGF-1 is inserted into a suitable expression system, such as Escherichia coli (E. coli). The bacteria are then cultured, and the recombinant protein is harvested and purified. This process ensures a high yield of LR3 IGF-1 with consistent quality and activity .

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.