IGF1 Human, A67T

Insulin Like Growth Factor-1, Mutant A67T Human Recombinant
Shipped with Ice Packs
In Stock

Description

IGF1 A67T Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 70 amino acids and having a molecular mass of Approximately 7.7 kDa.

The IGF1 A67T is purified by proprietary chromatographic techniques.

Product Specs

Introduction
The somatomedins, or insulin-like growth factors (IGFs), are a family of peptides involved in mammalian growth and development. IGF1 is a key mediator of growth hormone (GH) growth-promoting effects. Early research revealed that growth hormone did not directly promote sulfate incorporation into cartilage but instead operated through a serum component known as 'sulfation factor,' later renamed 'somatomedin'. Three primary somatomedins have been identified: somatomedin C (IGF1), somatomedin A (IGF2), and somatomedin B.
Description
IGF1 A67T Human Recombinant, produced in E. coli, is a single, non-glycosylated polypeptide chain containing 70 amino acids with an approximate molecular mass of 7.7 kDa. The purification process of IGF1 A67T involves proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
IGF1 A67T is lyophilized from a 0.2 µm filtered concentrated solution in 20 mM PB and 150 mM NaCl, at a pH of 6.0.
Solubility
To reconstitute the lyophilized IGF1 A67T, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can be further diluted into other aqueous solutions as needed.
Stability
For short-term storage (2-4 weeks), store the vial at 4°C. For extended storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 95.0% as determined by SDS-PAGE.
Synonyms
Somatomedin C, IGF-I, IGFI, IGF1, IGF-IA, Mechano growth factor, MGF.
Source
Escherichia Coli.
Amino Acid Sequence

GPETLCGAEL VDALQFVCGD RGFYFNKPTG YGSSSRRAPQ TGIVDECCFR SCDLRRLEMY CAPLKPTKSA.

Q&A

What is the A67T variant of human IGF1 and how prevalent is it?

The A67T variant represents a specific polymorphism in human insulin-like growth factor 1 where alanine at position 67 is replaced by threonine. This substitution occurs in the C-terminal region of the protein, which is a common location for IGF1 variants . According to comprehensive screening studies, IGF1 variants collectively occur in approximately 0.45% of the clinical population, which translates to about 4,508 occurrences per million people . The majority of detected variants (98%) have amino acid substitutions located at the C-terminus, including A67T along with other variants such as A62T, P66A, A67S, A67V, and A70T . The A67T variant is specifically distinguished from other similar variants through tandem mass spectrometry (MS/MS) analysis, requiring specialized detection methods beyond standard protein analysis techniques . Understanding the prevalence of this variant is essential for researchers investigating IGF1-related disorders and potential population-specific effects.

How does IGF1 function in normal human physiology and how might the A67T variant alter this?

IGF1 is a critical 70-amino acid peptide that mediates many biological effects of growth hormone (GH) in humans and other species . In normal physiology, GH stimulates IGF1 gene transcription through the transcription factor STAT5b, leading to the production of IGF1 mRNAs and subsequent synthesis of the protein . Most circulating IGF1 is bound to IGF-binding proteins (IGFBPs), with IGFBP-3 being the most abundant, which renders IGF1 inactive until needed . The protein exerts its actions by binding to the high-affinity IGF1 receptor (IGF1R) on cell surfaces, initiating complex intracellular signaling cascades including phosphorylation of insulin receptor substrate molecules and activation of PI3K-Akt and MAPK pathways . These pathways regulate numerous downstream effectors that influence autophagy, growth, stress resistance, oxidative stress response, and potentially lifespan . The A67T variant, being located in the C-terminal region, may alter IGF1's binding properties to its receptor and/or binding proteins, potentially affecting its activity and half-life in circulation . This structural change could impact the protein's ability to activate downstream signaling pathways, ultimately modifying IGF1's physiological effects throughout the body.

What methodologies are currently used to detect the A67T variant in clinical and research settings?

Detection of the A67T variant relies primarily on sophisticated mass spectrometry-based approaches. The variant is initially identified based on its predicted mass-to-charge ratios during liquid chromatography-mass spectrometry (LC-MS) analysis . While many IGF1 variants can be distinguished by their isotopic distribution and relative retention times during chromatographic separation, A67T specifically requires tandem mass spectrometry (MS/MS) for definitive identification and differentiation from other similar variants . Current high-throughput workflows for IGF1 quantification can process over 1,000 samples per day, utilizing automated pipetting robots and multiple MALDI-TOF-MS instruments working in parallel . Following initial detection at the protein level, confirmation typically involves de-identification of patient specimens and DNA sequencing to verify the underlying genetic polymorphism . Researchers should note that standard immunoassays for IGF1 may not distinguish between wild-type and variant forms, potentially leading to inaccurate quantification in individuals carrying the A67T variant. For comprehensive analysis, specialized antibodies may be employed during sample preparation, such as affinity-purified polyclonal rabbit anti-human IGF1 antibodies, followed by mass spectrometric analysis .

How does the A67T substitution potentially affect IGF1's molecular structure and interaction profile?

The A67T substitution introduces a threonine residue with a hydroxyl group in place of alanine's non-polar methyl group at position 67, likely creating subtle but potentially significant changes to IGF1's tertiary structure. This modification occurs in the C-terminal region, which has been implicated in interactions with IGF-binding proteins that regulate IGF1 bioavailability and half-life in circulation . Research has demonstrated that chromatographic peak areas of some IGF1 variants differ from that of the wild-type IGF1 present in the same patient, suggesting altered physical or chemical properties that affect their behavior during separation techniques . The positioning of A67T in or close to the C-domain may be particularly consequential, as variants in this region have been suggested to potentially have pathogenic effects . From a structural biology perspective, the introduction of the hydroxyl group could create new hydrogen bonding opportunities, potentially altering the protein's folding dynamics or surface properties. These structural changes may modify the binding kinetics between IGF1 and its receptor (IGF1R) or its various binding proteins, consequently affecting downstream signaling cascade activation including the PI3K-Akt and MAPK pathways that regulate numerous cellular processes .

What experimental approaches should researchers consider when studying the functional consequences of the A67T variant?

Researchers investigating the A67T variant should implement a multi-faceted experimental approach that combines biochemical, cellular, and potentially in vivo studies. Initial biochemical characterization should include protein binding assays comparing the affinity of wild-type and A67T IGF1 for the IGF1 receptor and various IGF-binding proteins, particularly IGFBP-3 . Surface plasmon resonance or isothermal titration calorimetry can provide quantitative binding parameters revealing potential alterations in association or dissociation rates. Cellular studies should examine downstream signaling activation, focusing on phosphorylation events in the PI3K-Akt and MAPK pathways using phospho-specific antibodies in Western blotting or ELISA assays . Researchers might generate cell lines expressing either wild-type IGF1, A67T variant, or both (to mimic heterozygosity) using CRISPR-Cas9 gene editing technology. Functional readouts should include assessments of cellular growth, survival under stress conditions, and autophagy regulation, as these processes are known to be influenced by IGF1 signaling . For in vivo studies, researchers might consider developing knock-in mouse models carrying the A67T substitution to examine systemic effects on growth, metabolism, and aging-related phenotypes. Throughout these studies, researchers must remember that heterozygosity is the most common presentation in human subjects, necessitating experimental designs that account for the presence of both wild-type and variant forms .

What is known about the potential impact of A67T on IGF1's role in aging and age-related diseases?

The potential impact of the A67T variant on IGF1's complex role in aging and age-related diseases represents an intriguing yet largely unexplored research frontier. IGF1 signaling has been characterized as having dual "Jekyll and Hyde" characteristics in the aging brain and other tissues, with both beneficial and detrimental effects depending on context . In neurodegenerative disorders like Alzheimer's disease, reducing IGF1 signaling may be protective by enhancing clearance of pathological proteins and maintaining cellular homeostasis . Conversely, in cerebrovascular disease and acute neuronal injury scenarios, IGF1 appears beneficial for neuronal recovery and vascular health . The A67T variant, by potentially altering IGF1's binding affinities and signaling capacities, could shift this delicate balance in specific ways that remain to be fully characterized. Given that variants in or close to the C-domain (which includes A67T) may have pathogenic potential, this substitution might influence age-related disease susceptibility or progression . Research examining the correlation between A67T carrier status and trajectories of cognitive decline, cerebrovascular health, or neurodegenerative disease incidence would be particularly valuable. Additionally, investigating whether the A67T variant alters IGF1 resistance, which has been reported to occur in aging and diseased brains, could provide insights into its role in age-related pathologies .

How does the A67T variant compare to other known IGF1 variants in terms of functional significance?

The A67T variant exists within a spectrum of IGF1 variants that have been identified in human populations, each with potentially distinct functional implications. Cross-species sequence comparison has been used to evaluate the potential pathogenicity of different variants, with some C-terminal variants showing varying degrees of potential pathogenicity . Unlike variants in the B-domain (residues 3-29) that might directly affect receptor binding, C-terminal variants like A67T may predominantly influence interactions with IGF-binding proteins or alter protein stability in circulation . Interestingly, some isobaric variants like A38V and A67V show age-dependent frequency differences, being detected more frequently in children than in adults, suggesting potential developmental stage-specific effects that might also apply to A67T . When comparing the seven identified IGF1 variants located at the C-terminus (A62T, P66A, A67S, A67V, A67T, and A70T), researchers should consider their relative frequencies, structural impacts, and functional consequences . Additionally, six previously unreported variants have been identified (Y31H, S33P, T41I, R50Q, R56K, and A62T), expanding the landscape of IGF1 variation that provides context for understanding A67T . Comprehensive comparative studies examining how different variants affect IGF1's binding properties, signaling activation, and biological outcomes would significantly advance our understanding of structure-function relationships in this critical growth factor and help position A67T within this broader functional landscape.

What are the challenges in developing accurate detection and quantification methods specifically for the A67T variant?

Developing accurate detection and quantification methods for the A67T variant presents several significant challenges that researchers must address. First, the subtle nature of the amino acid substitution (alanine to threonine) results in a small mass difference that can be difficult to resolve using standard mass spectrometry approaches, necessitating high-resolution instruments and specialized methods like tandem mass spectrometry (MS/MS) for definitive identification . Second, the variant's similarity to wild-type IGF1 may result in cross-reactivity with antibodies used in immunoassays or immunocapture steps, potentially leading to inaccurate quantification. Third, the heterozygous nature of most A67T presentations means that both wild-type and variant forms coexist in patient samples, requiring methods that can accurately distinguish and quantify both forms simultaneously . Fourth, the binding of IGF1 to IGF-binding proteins in circulation complicates sample preparation, necessitating effective techniques to release IGF1 from these complexes without affecting the protein's structure or the ability to detect the variant . Additionally, the chromatographic behavior of the A67T variant may differ from wild-type IGF1, requiring optimization of separation parameters to ensure accurate quantification . Researchers developing such methods should implement a rigorous validation process focusing on specificity, sensitivity, accuracy, and precision specifically for detecting the A67T variant, ideally using confirmed A67T samples and suitable internal standards for quantification.

How should researchers approach the development of in vitro and in vivo models to study A67T effects?

Developing appropriate experimental models to study the A67T variant requires careful consideration of multiple factors to ensure physiological relevance. For in vitro models, researchers should consider generating cell lines expressing either wild-type IGF1, A67T variant, or both (to mimic heterozygosity) using site-directed mutagenesis or CRISPR-Cas9 gene editing . Hepatocytes would be particularly relevant as they represent the primary source of circulating IGF1, though cell types from target tissues (neurons, myocytes, osteoblasts) should also be considered to examine tissue-specific effects . These cellular models should be used to assess differences in IGF1 secretion, stability, receptor binding, and downstream signaling activation between wild-type and A67T variants. For in vivo approaches, knock-in mice with the A67T substitution in the endogenous IGF1 gene would provide a system for studying systemic effects on growth, metabolism, tissue development, and aging . Both heterozygous and homozygous models should be developed to reflect the genetic diversity observed in human populations . Additionally, conditional expression systems could help control the timing and tissue specificity of A67T expression, allowing for more nuanced studies of developmental or tissue-specific effects. Humanized mouse models expressing human IGF1 with the A67T variant might provide even greater translational relevance. Phenotypic assessments should be comprehensive, including growth curves, metabolic parameters, tissue-specific IGF1 actions, responses to various stressors, and age-related changes to fully characterize the variant's impact across different physiological contexts and life stages.

What implications does the A67T variant have for IGF1-targeted therapies in various disease contexts?

The A67T variant could have significant implications for the development and application of IGF1-targeted therapies across various disease contexts. Understanding how this variant affects IGF1's binding properties and signaling capacity is crucial for predicting therapeutic responses in carriers. In neurodegenerative diseases like Alzheimer's disease, where IGF1 has demonstrated dual roles, the A67T variant might alter the balance between beneficial and detrimental effects . For instance, if A67T enhances IGF1's ability to promote clearance of pathological proteins, carriers might respond differently to therapies targeting IGF1 signaling compared to individuals with wild-type IGF1. In cerebrovascular disease and acute neuronal injury, where IGF1 appears beneficial for neuronal recovery and vascular health, the variant might influence the efficacy of IGF1-based neuroprotective strategies . Evidence from animal models showing that IGF1 administration improves functional recovery and enhances neurogenesis after ischemic injury suggests potential therapeutic applications that could be affected by variant-induced changes in IGF1 function . Additionally, in Parkinson's disease, where elevated IGF1 levels have been associated with early-stage disease and potentially worse motor outcomes, the A67T variant's effect on IGF1 activity might influence disease progression and treatment response . For precision medicine approaches, identifying patients with the A67T variant before initiating IGF1-targeted therapies could allow for personalized dosing regimens or alternative treatment strategies. Future clinical trials involving IGF1-modulating therapies should consider screening for the A67T variant to assess whether carrier status influences treatment outcomes, potentially leading to more tailored therapeutic approaches based on individual genetic profiles.

What key experiments would advance our understanding of the A67T variant's structural and functional impact?

Advancing our understanding of the A67T variant requires a strategic experimental approach addressing several key knowledge gaps. First, high-resolution structural studies using X-ray crystallography or cryo-electron microscopy comparing wild-type and A67T IGF1 would provide critical insights into any conformational changes induced by the substitution, particularly focusing on regions involved in receptor and binding protein interactions . Hydrogen-deuterium exchange mass spectrometry could complement these studies by revealing dynamic structural differences between the variants. Second, comprehensive binding kinetics studies using surface plasmon resonance or biolayer interferometry should examine interactions between A67T and all major binding partners (IGF1R, insulin receptor, and various IGFBPs), measuring association and dissociation rates to quantify any affinity changes . Third, cellular signaling experiments in relevant cell types (hepatocytes, neurons, myocytes) comparing pathway activation dynamics between wild-type and A67T variants would illuminate functional consequences, ideally using phosphoproteomics to capture the full spectrum of signaling differences . Fourth, isotope labeling studies in appropriate animal models could assess whether the A67T variant exhibits altered half-life or tissue distribution compared to wild-type IGF1. Fifth, gene editing to introduce the A67T variant in pluripotent stem cells followed by differentiation into various lineages would allow examination of developmental and cell type-specific effects. Finally, computational approaches including molecular dynamics simulations could predict how the threonine substitution affects protein flexibility, solvent accessibility, and interaction energetics with binding partners, generating hypotheses for experimental validation.

How might the A67T variant contribute to our broader understanding of IGF1 biology and personalized medicine?

The A67T variant offers a unique window into IGF1 biology that could significantly enhance our understanding of structure-function relationships in this critical growth factor and inform personalized medicine approaches. By studying how this specific amino acid substitution affects IGF1's interactions and signaling, researchers can gain insights into the precise structural determinants of IGF1's diverse biological functions . This knowledge could help resolve the paradoxical "Jekyll and Hyde" nature of IGF1 in different contexts, particularly in aging and disease processes . The variant's location in the C-terminal region highlights the importance of this often-overlooked domain in mediating IGF1's biological effects, potentially leading to refined models of IGF1 action that account for region-specific functions . From a personalized medicine perspective, characterizing the A67T variant's effects could establish a paradigm for how IGF1 genetic variation influences disease susceptibility, progression, and treatment response across conditions ranging from growth disorders to neurodegenerative diseases and cancer . This could lead to genetic screening recommendations for certain at-risk populations and tailored therapeutic approaches based on IGF1 variant status. Additionally, understanding the A67T variant could inform the development of modified IGF1 proteins with enhanced therapeutic properties, such as increased specificity for certain receptors or altered binding to IGFBPs for improved pharmacokinetics. Ultimately, detailed characterization of the A67T variant and similar polymorphisms could transition IGF1-related therapies from a one-size-fits-all approach to precision interventions optimized for individual genetic profiles.

What methodological innovations would facilitate large-scale screening and functional characterization of the A67T variant?

Advancing large-scale screening and functional characterization of the A67T variant requires innovations across multiple methodological domains. In detection technology, developing simplified mass spectrometry approaches that maintain high specificity for the A67T variant while increasing throughput and reducing cost would facilitate population-level screening . This might include targeted MS/MS methods optimized specifically for the A67T variant or novel immunocapture approaches using antibodies with differential affinities for wild-type versus variant IGF1 . For high-throughput functional characterization, multiplexed cellular assays using reporter systems to simultaneously monitor multiple IGF1 signaling pathways could rapidly assess the variant's impact across different cellular contexts. Microfluidic organ-on-a-chip platforms incorporating cells expressing either wild-type or A67T variant could provide physiologically relevant systems for functional studies while reducing animal use. Computationally, developing machine learning algorithms trained on existing IGF1 variant data could help predict the functional impact of newly discovered variants and prioritize them for experimental validation. For clinical correlation studies, establishing biobanks specifically collecting samples from individuals with confirmed IGF1 variants along with detailed phenotypic data would create valuable resources for retrospective and prospective studies. Additionally, creating standardized protocols for IGF1 variant detection and quantification would enable data sharing and meta-analyses across different research groups, accelerating knowledge accumulation. Finally, international research collaborations and consortia focused specifically on IGF1 variants could coordinate efforts to systematically characterize their prevalence, functional impacts, and clinical correlations across diverse populations, maximizing research efficiency and impact.

Product Science Overview

Introduction

Insulin-Like Growth Factor-1 (IGF-1) is a protein that plays a crucial role in growth and development, particularly during childhood. It is structurally similar to insulin and is part of the insulin gene family. IGF-1 is produced primarily in the liver and acts as an endocrine hormone, but it is also synthesized in various tissues in a paracrine/autocrine manner . The mutant A67T variant of IGF-1 refers to a specific alteration in the amino acid sequence where alanine (A) at position 67 is replaced by threonine (T).

Biological Significance

IGF-1 is a potent mitogen that stimulates the proliferation and survival of various cell types, including muscle, bone, and cartilage tissues . It mediates many of the growth-promoting effects of growth hormone (GH) and is involved in cellular differentiation, transformation, and apoptosis. The A67T mutation in IGF-1 has been studied for its potential impact on the protein’s function and its implications in various diseases and therapeutic applications.

Preparation Methods

Recombinant human IGF-1, including the A67T mutant, is typically produced using bacterial expression systems such as Escherichia coli . The gene encoding the IGF-1 protein is inserted into a plasmid vector, which is then introduced into the bacterial cells. The bacteria are cultured, and the recombinant protein is expressed and subsequently purified through various chromatographic techniques. The final product is often lyophilized for stability and ease of storage.

Chemical Reactions and Analysis

The A67T mutation in IGF-1 can affect the protein’s structure and function. Analytical techniques such as mass spectrometry, nuclear magnetic resonance (NMR) spectroscopy, and X-ray crystallography are employed to study the structural changes induced by the mutation . Functional assays, including cell proliferation and receptor binding studies, are conducted to assess the biological activity of the mutant protein compared to the wild-type IGF-1.

Applications and Implications

Recombinant IGF-1, including the A67T mutant, has significant therapeutic potential. It has been investigated for its role in improving insulin sensitivity and glycemic control in patients with type II diabetes . Additionally, IGF-1 is being explored for its potential in treating growth deficiencies, muscle wasting diseases, and certain cancers. The A67T mutation, in particular, may offer insights into the protein’s function and its role in disease mechanisms.

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.