Insulin Human, His

Insulin Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Insulin Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 109 amino acids (25-110 a.a) and having a molecular mass of 11.8kDa. Insulin is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Insulin is a hormone that plays a crucial role in regulating blood sugar levels. It facilitates the entry of glucose into cells, promoting its utilization for energy production and storage. Additionally, insulin stimulates the synthesis of glycogen in the liver, further contributing to glucose homeostasis.
Description
This recombinant insulin, produced in E.Coli, is a single-chain polypeptide consisting of 109 amino acids (specifically, amino acids 25-110). It has a molecular weight of 11.8kDa and lacks glycosylation. For purification purposes, a 23 amino acid His-tag is attached to the N-terminus of the insulin molecule. The purification process involves proprietary chromatographic techniques.
Physical Appearance
The product is a clear solution that has undergone sterile filtration.
Formulation
The insulin protein solution is provided at a concentration of 0.5mg/ml. The solution contains Phosphate-Buffered Saline (pH 7.4) and 10% Glycerol.
Stability
For optimal storage, refrigeration at 4°C is recommended if the entire vial will be used within 2-4 weeks. For extended storage periods, the product should be frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freezing and thawing cycles should be avoided.
Purity
The purity of the insulin is determined by SDS-PAGE analysis and is greater than 95.0%.
Biological Activity
The biological activity of the insulin is evaluated through a cell proliferation assay using MCF7 human breast cancer cells. The ED50, which represents the concentration of insulin required to achieve half-maximal cell proliferation, is less than or equal to 4 ug/ml.
Synonyms

Insulin, Insulin-Dependent Diabetes Mellitus 2, Preproinsulin, Proinsulin, MODY10, IDDM1, IDDM2, IDDM, ILPR, IRDN.    

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSFVNQHLC GSHLVEALYL VCGERGFFYT PKTRREAEDL QVGQVELGGG PGAGSLQPLA LEGSLQKRGI VEQCCTSICS LYQLENYCN

Q&A

What is the molecular structure of human insulin and its significance in experimental research?

Human insulin is a peptide hormone consisting of 51 amino acids organized into two chains: an A chain (21 amino acids) and a B chain (30 amino acids) connected by disulfide bonds . The precise sequence of the A chain is GIVEQCCTSICSLYQLENYCN, while the B chain sequence is FVNQHLCGSHLVEALYLVCGERGFFYTPKT . The molecular formula is C257H383N65O77S6 with an average weight of 5808.0 Da .

The three-dimensional structure of insulin is critical for its biological activity, particularly the areas that interact with the insulin receptor. In experimental research, understanding this structure is fundamental because:

  • The tertiary structure determines receptor binding affinity and specificity

  • Disulfide bond integrity affects stability and biological activity

  • The C-terminal region of the B-chain undergoes conformational changes during receptor binding

  • Small modifications in the amino acid sequence can significantly alter pharmacokinetic properties

Researchers should note that the structure-function relationship makes human insulin an excellent model for studying protein-receptor interactions and for designing insulin analogs with modified properties for specific experimental purposes .

What are the primary mechanisms through which human insulin regulates cellular metabolism?

Human insulin regulates cellular metabolism through several interconnected pathways that researchers should consider in experimental design:

Primary Signal Transduction Pathway:

  • Insulin binds to the insulin receptor (IR), a heterotetrameric protein with two extracellular α-subunits and two transmembrane β-subunits

  • Binding activates the tyrosine kinase activity of the β-subunits, initiating autophosphorylation

  • The activated receptor phosphorylates intracellular substrates including insulin receptor substrates (IRS), Cbl, APS, Shc, and Gab 1

  • These activated proteins lead to downstream activation of PI3 kinase and Akt

  • Akt regulates glucose transporter 4 (GLUT4) and protein kinase C (PKC), which play critical roles in metabolism

Metabolic Effects in Target Tissues:

TissuePrimary Insulin ActionsExperimental Considerations
Skeletal MuscleGlucose uptake via GLUT4 translocation; glycogen synthesis; protein synthesisConstitutes ~70% of insulin-stimulated glucose disposal; key tissue for studying insulin resistance
Adipose TissueGlucose uptake; lipogenesis; inhibition of lipolysisImportant for studying metabolic syndrome and obesity-related insulin resistance
LiverInhibition of gluconeogenesis; promotion of glycogen synthesis; lipogenesisCentral for glucose homeostasis research; exhibits direct and indirect insulin effects
BrainEnhanced learning and memory; regulation of appetiteIncreasingly recognized as an insulin-responsive tissue with implications for cognitive research

For rigorous experimental design, researchers should account for the tissue-specific nature of insulin signaling and the complex crosstalk between metabolic pathways .

How is insulin secretion regulated in experimental models?

Understanding insulin secretion regulation is essential for designing experiments that accurately reflect physiological conditions:

Glucose-Dependent Regulation:

  • Threshold effect: Insulin secretion is initiated at glucose concentrations above 5 mM, while insulin biosynthesis occurs at lower concentrations (2-4 mM)

  • Beta cells sense glucose through metabolism-dependent mechanisms involving KATP channels

  • Glucose metabolism increases ATP:ADP ratio, closing KATP channels and depolarizing the cell membrane

  • Depolarization triggers calcium influx through voltage-dependent calcium channels, stimulating insulin exocytosis

Incretin-Mediated Regulation:

  • Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) enhance glucose-stimulated insulin secretion

  • These incretin hormones bind to G-protein-coupled receptors on β-cell membranes

  • This increases cellular cAMP levels and potentiates glucose-stimulated insulin secretion

  • Incretin action is independent of KATP channel closure, providing an additional regulatory pathway

Other Regulatory Factors:

  • Amino acids (particularly arginine and leucine) stimulate insulin secretion

  • Neural regulation via parasympathetic and sympathetic inputs

  • Hormonal modulation by glucagon, somatostatin, and stress hormones

When designing experiments involving insulin secretion, researchers should carefully consider:

  • The glucose concentration used (fasting vs. stimulatory levels)

  • The presence or absence of incretins in the experimental system

  • The potential influence of neural and hormonal factors

  • The duration of stimulation (acute vs. chronic exposure)

What standardized methodologies exist for measuring insulin activity in biological systems?

Researchers investigating insulin require reliable methods to measure insulin activity:

In Vitro Assays:

  • Receptor Binding Assays:

    • Competitive binding assays using radiolabeled insulin and cell lines expressing insulin receptors

    • Surface plasmon resonance for real-time binding kinetics

    • FRET-based approaches for investigating conformational changes

  • Signaling Pathway Activation:

    • Western blotting for phosphorylation of insulin receptor and downstream targets (IRS, Akt)

    • Phospho-specific ELISA for quantitative measurement of signaling activation

    • Immunofluorescence for spatial distribution of signaling events

    • Live-cell imaging with fluorescent biosensors for real-time pathway activation

  • Metabolic Readouts:

    • Glucose uptake assays using radiolabeled or fluorescent glucose analogs

    • Glycogen synthesis measurement

    • Lipogenesis assays measuring incorporation of labeled acetate or glucose

    • Protein synthesis determination using labeled amino acids

Ex Vivo and In Vivo Methods:

  • Islet Perifusion Studies:

    • Real-time measurement of insulin secretion from isolated islets

    • Allows evaluation of dynamic insulin secretion in response to various stimuli

  • Hyperinsulinemic-Euglycemic Clamp:

    • Gold standard for assessing insulin sensitivity in vivo

    • Measures the glucose infusion rate required to maintain euglycemia during insulin infusion

  • Insulin Tolerance Test:

    • Measures blood glucose decrease following insulin administration

    • Provides a practical index of whole-body insulin sensitivity

When selecting methodologies, researchers should consider the specific aspect of insulin action they are investigating and choose appropriate positive and negative controls to ensure assay validity .

How do insulin's pleiotropic effects impact experimental design in complex physiological research?

Insulin's effects extend beyond glucose metabolism, creating challenges and opportunities for complex physiological research:

Cardiovascular Effects:

  • Insulin influences vascular compliance and endothelial function

  • Promotes vasodilation through nitric oxide production

  • Affects sodium reabsorption in the kidneys, influencing blood pressure regulation

Neurological Functions:

  • Enhances learning and memory, particularly verbal memory

  • Contributes to thermoregulatory and glucoregulatory responses to food intake

  • Impacts central nervous system function through specific receptors in various brain regions

Reproductive System Effects:

  • Stimulates gonadotropin-releasing hormone from the hypothalamus

  • Influences fertility and reproductive function

  • Interacts with sex hormones in metabolic regulation

Methodological Considerations for Researchers:

Research AreaConfounding FactorsRecommended Controls
Metabolism StudiesNon-metabolic insulin effects altering energy homeostasisTime-matched experiments; specific pathway inhibitors
Cardiovascular ResearchDirect vascular effects vs. metabolic improvementsPathway-specific interventions; hyperinsulinemic-euglycemic clamps
Neuroscience InvestigationsCentral vs. peripheral insulin actionsBrain-specific insulin receptor manipulations; intranasal insulin administration
Reproductive StudiesInteraction with sex hormonesSex-stratified analyses; gonadectomy models with hormone replacement

Researchers should implement experimental designs that can distinguish direct insulin effects from indirect consequences of improved metabolism. This might include:

  • Using specific insulin receptor antagonists

  • Employing tissue-specific insulin receptor knockout models

  • Comparing insulin to non-insulin glucose-lowering interventions

  • Implementing time-course studies to separate rapid signaling from transcriptional effects

What are the molecular mechanisms underlying insulin resistance and how can they be accurately modeled in research?

Insulin resistance represents a major research area with important methodological considerations:

Molecular Mechanisms:

  • Receptor-Level Defects:

    • Decreased insulin receptor expression

    • Impaired insulin binding

    • Reduced receptor autophosphorylation

  • Post-Receptor Signaling Defects:

    • Increased serine/threonine phosphorylation of IRS proteins

    • Reduced PI3K activation and PIP3 generation

    • Impaired Akt phosphorylation and activation

    • Defective GLUT4 translocation

  • Inflammatory Mechanisms:

    • Activation of inflammatory kinases (JNK, IKK, PKC isoforms)

    • Increased cytokine signaling interfering with insulin pathways

    • Macrophage infiltration in metabolic tissues

  • Lipid-Mediated Mechanisms:

    • Diacylglycerol activation of novel PKC isoforms

    • Ceramide inhibition of Akt

    • Altered membrane lipid composition affecting receptor function

Experimental Models for Insulin Resistance:

Model TypeAdvantagesLimitationsMethodological Considerations
Cell CulturePrecise molecular manipulation; high throughputLacks systemic factors; acute rather than chronicUse physiological insulin concentrations (0.1-10 nM); include appropriate time courses
Palmitate-InducedMimics lipotoxicity; rapid inductionMay not reflect all aspects of insulin resistanceControl fatty acid composition and albumin binding; monitor cell viability
Hyperinsulinemia-InducedReflects physiological downregulationCan be difficult to distinguish from toxicityUse pulsatile rather than constant insulin exposure
Genetic Models (in vivo)Tissue-specific manipulation possibleCompensatory mechanisms may developInducible systems can reduce developmental adaptations
Diet-Induced (in vivo)Closely mimics human pathophysiologyStrain-dependent responses; long induction timesControl for changes in body composition; pair-feeding may be necessary

When designing insulin resistance studies, researchers should:

  • Validate insulin resistance using multiple readouts (signaling, glucose uptake, metabolic effects)

  • Consider tissue-specific differences in insulin resistance mechanisms

  • Account for the time-dependent progression of insulin resistance

  • Include controls for confounding factors like inflammation, oxidative stress, and ER stress

How can researchers effectively distinguish between genomic and non-genomic actions of insulin in experimental systems?

Insulin exerts both rapid non-genomic effects and longer-term genomic effects, presenting methodological challenges:

Characteristics of Genomic vs. Non-Genomic Actions:

ParameterGenomic ActionsNon-Genomic Actions
Time CourseHours (typically >2h)Seconds to minutes (<30 min)
MechanismsTranscriptional regulationProtein phosphorylation; vesicle translocation
InhibitionBlocked by transcription/translation inhibitorsResistant to transcription/translation inhibitors
Concentration RequiredOften effective at physiological concentrationsMay require higher concentrations for some effects
Receptor DependenceTypically canonical insulin receptor-dependentMay involve insulin receptor or alternative receptors/pathways

Methodological Approaches:

  • Temporal Discrimination:

    • Perform detailed time-course experiments from seconds to hours

    • Compare rapid effects (glucose transport, enzyme activity) with delayed responses (protein synthesis, gene expression)

    • Use pulse-chase experimental designs to separate initial signaling from downstream consequences

  • Pharmacological Tools:

    • Apply transcription inhibitors (actinomycin D) or translation inhibitors (cycloheximide) to block genomic effects

    • Use pathway-specific inhibitors to dissect signaling cascades

    • Employ receptor antagonists to determine receptor specificity

  • Genetic Approaches:

    • Utilize cells with mutated insulin receptors that maintain non-genomic signaling but lack genomic effects

    • Implement CRISPR-Cas9 to modify specific signaling nodes

    • Develop inducible expression systems for controlled temporal studies

  • Advanced Techniques:

    • Real-time imaging with fluorescent biosensors for immediate signaling events

    • Chromatin immunoprecipitation to detect insulin-regulated transcription factor binding

    • Ribosome profiling to assess translational effects

    • Phosphoproteomics to capture early signaling events comprehensively

Researchers should design experiments with appropriate controls to account for:

  • Potential crosstalk between genomic and non-genomic pathways

  • Tissue-specific variations in insulin action mechanisms

  • Concentration-dependent effects that may engage different pathways

  • The influence of experimental conditions (cell confluency, serum starvation, etc.) on insulin responsiveness

What methodological approaches best address the contradictions in insulin signaling data between different experimental models?

Researchers frequently encounter contradictory findings when studying insulin signaling across different models:

Common Sources of Data Contradictions:

  • Model-Specific Differences:

    • Species variations in insulin signaling components

    • Differences between primary cells and immortalized cell lines

    • Variations in receptor/pathway component expression levels

    • Altered metabolic states of different models

  • Methodological Variations:

    • Differences in insulin concentrations used (physiological vs. pharmacological)

    • Varied duration of insulin stimulation

    • Diverse cell culture conditions (glucose concentration, serum components)

    • Different analytical techniques and their sensitivity/specificity

Systematic Approaches to Resolve Contradictions:

  • Cross-Model Validation:

    • Systematically test hypotheses across multiple models under standardized conditions

    • Include appropriate positive and negative controls for each model

    • Directly compare primary cells to cell lines when possible

    • Validate in vitro findings in ex vivo or in vivo systems

  • Standardization of Methodology:

    • Establish dose-response relationships for insulin in each model

    • Perform detailed time-course studies to capture both early and late responses

    • Control for cell density, passage number, and culture conditions

    • Normalize signaling responses to receptor expression levels

  • Comprehensive Pathway Analysis:

    • Examine multiple nodes within signaling pathways rather than single endpoints

    • Utilize phospho-specific antibodies to assess activation states

    • Implement mass spectrometry-based phosphoproteomics for unbiased assessment

    • Consider pathway crosstalk and compensatory mechanisms

  • Advanced Statistical Approaches:

    • Apply meta-analysis techniques to integrate findings across studies

    • Use multivariate analysis to identify covariates affecting insulin responses

    • Implement Bayesian approaches to incorporate prior knowledge

    • Develop mathematical models to predict context-dependent signaling outcomes

To effectively address contradictions, researchers should:

  • Clearly report all experimental conditions in publications

  • Include detailed methods sections with critical parameters

  • Consider biological context when interpreting results

  • Acknowledge limitations of specific models

  • Design experiments that directly test alternative hypotheses

How can researchers effectively study insulin's role in integrating multi-tissue metabolic responses?

Insulin coordinates metabolism across multiple tissues, presenting unique research challenges:

Tissue-Specific Insulin Actions and Their Integration:

TissuePrimary Insulin ActionsIntegration with Other Tissues
LiverSuppresses gluconeogenesis; promotes glycogen synthesis and lipogenesisReleases glucose/VLDL to fuel peripheral tissues; responds to muscle-derived amino acids
Skeletal MuscleStimulates glucose uptake and glycogen synthesis; promotes protein synthesisReleases amino acids during insulin deficiency; lactate production affects liver metabolism
Adipose TissuePromotes glucose uptake and lipogenesis; inhibits lipolysisReleases or stores FFAs affecting muscle/liver insulin sensitivity; secretes adipokines
BrainRegulates appetite and autonomic outputs; affects cognitionControls neural signals to liver, muscle, and adipose; regulates counter-regulatory hormone release
PancreasSuppresses glucagon secretion from α cellsDetermines insulin:glucagon ratio affecting all other tissues

Methodological Approaches:

  • Integrated In Vivo Techniques:

    • Hyperinsulinemic-euglycemic clamps with tissue-specific glucose uptake measurements

    • Arterio-venous difference measurements across tissue beds

    • Stable isotope tracer studies to track metabolic fluxes

    • Metabolic cages for continuous assessment of whole-body metabolism

  • Ex Vivo Organ Crosstalk:

    • Conditioned media experiments transferring secreted factors between tissues

    • Co-culture systems with multiple cell types

    • Microfluidic "organ-on-chip" approaches with connected tissue compartments

    • Perifusion systems for dynamic secretion studies

  • In Vivo Tissue-Specific Manipulations:

    • Tissue-specific insulin receptor knockout models

    • Viral vector-mediated gene delivery to specific tissues

    • Inducible, tissue-specific transgene expression

    • Selective inhibition of tissue-specific insulin action using antisense oligonucleotides

  • Systems Biology Approaches:

    • Multi-omics integration (transcriptomics, proteomics, metabolomics)

    • Computational modeling of cross-tissue metabolic fluxes

    • Agent-based modeling of tissue interactions

    • Network analysis of hormone signaling integration

Researchers studying multi-tissue integration should:

  • Design studies that simultaneously assess multiple tissues

  • Consider the temporal sequence of insulin actions across tissues

  • Account for fed/fasted state differences in tissue insulin sensitivity

  • Recognize that perturbations in one tissue can have secondary effects on others

  • Integrate findings from reductionist models into a systems-level understanding of metabolism

What are the cutting-edge methods for investigating insulin's role beyond traditional metabolic pathways?

Recent research has expanded our understanding of insulin's functions beyond classical metabolic regulation:

Emerging Non-Traditional Roles of Insulin:

  • Neurocognitive Functions:

    • Enhances learning and memory processes

    • Contributes to hippocampal synaptic plasticity

    • Influences neurodegenerative disease pathways

    • Affects reward processing and feeding behavior

  • Immune System Modulation:

    • Regulates inflammatory cytokine production

    • Affects immune cell metabolism and function

    • Influences macrophage polarization

    • Impacts wound healing and tissue repair processes

  • Cellular Stress Responses:

    • Modulates endoplasmic reticulum stress

    • Influences autophagy and proteostasis

    • Affects cellular senescence pathways

    • Regulates mitochondrial dynamics and function

  • Epigenetic Regulation:

    • Alters DNA methylation patterns

    • Influences histone modifications

    • Regulates non-coding RNA expression

    • Contributes to metabolic memory phenomena

Advanced Methodological Approaches:

Research AreaCutting-Edge TechniquesMethodological Considerations
NeurocognitiveOptogenetic manipulation of insulin-responsive neurons; in vivo microdialysis with neurotransmitter measurement; functional neuroimaging during insulin administrationBrain-specific insulin delivery (e.g., intranasal); controlling for systemic metabolic effects
ImmunometabolicSingle-cell metabolic profiling of immune cells; isotope tracing in specific immune populations; spatial transcriptomics of tissue-resident immune cellsMaintaining physiological insulin concentrations; accounting for indirect effects through glucose modulation
Stress ResponsesLive-cell imaging of stress responses; proximity labeling of insulin-regulated stress proteins; stress-specific organoid modelsDistinguishing direct insulin effects from adaptive responses to metabolic changes
EpigeneticsChIP-seq for insulin-regulated transcription factors; ATAC-seq for chromatin accessibility; single-cell multi-omics approachesTemporal considerations (acute vs. chronic insulin exposure); tissue-specific epigenetic responses

For researchers exploring these frontier areas, recommended approaches include:

  • Combining targeted and unbiased screening approaches

  • Implementing single-cell technologies to address cellular heterogeneity

  • Developing tissue-specific insulin resistance models that preserve insulin action in tissues of interest

  • Utilizing conditional knockout strategies with temporal control

  • Applying systems biology approaches to integrate diverse datasets

  • Adapting methods from specialized fields (neuroscience, immunology, etc.) to insulin research questions

Product Science Overview

Background of Insulin (Human Recombinant, His Tag)

Insulin is a crucial hormone produced by the pancreas that regulates glucose levels in the blood. It facilitates the uptake of glucose into cells, thereby providing them with energy. Insulin deficiency or resistance leads to diabetes, a condition that affects millions worldwide.

Recombinant human insulin is a form of insulin that is produced using recombinant DNA technology. This method involves inserting the human insulin gene into bacteria or yeast, which then produce insulin that can be harvested and purified. This technology has revolutionized diabetes treatment by providing a consistent and reliable source of insulin.

His Tag (Histidine Tag) is a sequence of histidine residues added to proteins to facilitate their purification. The His tag binds to nickel ions, allowing the tagged protein to be separated from other cellular components using a technique called affinity chromatography.

Production of Insulin (Human Recombinant, His Tag)
  1. Gene Insertion: The human insulin gene is inserted into a plasmid, a small circular piece of DNA.
  2. Transformation: The plasmid is introduced into a host organism, typically Escherichia coli (E. coli) or yeast.
  3. Expression: The host organism expresses the insulin gene, producing insulin protein with a His tag.
  4. Purification: The His-tagged insulin is purified using affinity chromatography, where it binds to a nickel column and is separated from other proteins.
  5. Refolding: The purified insulin is refolded into its active form and further purified to ensure high quality and activity.
Applications and Benefits
  • Diabetes Treatment: Recombinant human insulin is used to treat diabetes, providing a reliable and consistent source of insulin for patients.
  • Research: His-tagged insulin is used in research to study insulin’s structure, function, and interactions with other molecules.
  • Biotechnology: The His tag facilitates the purification of insulin, making the production process more efficient and cost-effective.

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.