Insulin Antibody

Insulin, Mouse Anti-Human
Shipped with Ice Packs
In Stock

Description

Definition and Formation

Insulin antibodies are proteins generated when the immune system identifies insulin as a foreign antigen . They are classified into two categories:

  • Exogenous insulin antibodies: Develop in insulin-treated individuals, particularly with non-human insulin formulations (e.g., bovine, porcine) .

  • Endogenous insulin autoantibodies (IAAs): Found in insulin-naïve individuals, often linked to autoimmune disorders like type 1 diabetes or insulin autoimmune syndrome (IAS) .

Subclasses and Clinical Associations

Insulin antibodies belong to various immunoglobulin subclasses, each with distinct clinical implications:

SubclassPrevalenceClinical Correlation
IgGMost common (92/98 cases) Linked to insulin resistance and prolonged action
IgE24.32% of IA-positive Associated with injection-site reactions
IgM7/98 cases Correlated with hypoglycemia
IgARareLimited data on clinical impact

IgG1 and IgG4 are the predominant IgG isotypes, while IgE antibodies are implicated in allergic reactions to insulin .

Mechanisms of Dysglycemia

High-affinity antibodies bind insulin tightly, delaying its clearance and prolonging hypoglycemic effects. Low-affinity antibodies with high binding capacity act as reservoirs, causing erratic insulin release and glucose fluctuations . Key findings include:

  • Postprandial hyperglycemia: Antibody-bound insulin fails to act promptly after meals .

  • Delayed hypoglycemia: Late insulin release from antibody complexes .

  • Insulin resistance: Antibodies block insulin-receptor interaction .

Diagnostic Approaches

Laboratory methods to identify clinically significant antibodies:

MethodPurposeKey CriteriaReference
PEG precipitationDifferentiate antibody-bound vs. free insulin>50% decline in immunoreactivity post-PEG
Gel filtration chromatographyDetect high molecular weight immune complexesPresence of HMW insulin complexes
Radioligand binding assayQuantify antibody levelsThreshold: >0.4 U/mL (positive)

Inhaled Insulin and Antibody Development

A 24-week randomized trial compared inhaled insulin (INH) with subcutaneous insulin (SC) in type 1 diabetes:

Subclass-Specific Effects

A study of 516 type 2 diabetes patients on premixed insulin analogs found:

  • IgE-IA: Associated with elevated serum insulin (median Δ: +15.8 μIU/mL) and injection-site reactions (24.32%) .

  • IgM-IA: Linked to hypoglycemia (57.14% frequency) .

  • IgG-IA: Predominant subclass but no direct glycemic impact .

Immunomodulation Outcomes

In severe cases of antibody-mediated dysglycemia:

  • Rituximab: Reduced IA titers and stabilized glucose in 1/2 patients .

  • Plasmapheresis: Effective in acute settings to lower antibody-bound insulin .

Clinical Management Strategies

  • Insulin regimen adjustment: Switching to human or analog insulins reduces immunogenicity .

  • Immunosuppression: Reserved for refractory cases with labile glucose .

  • Monitoring: Regular HbA1c, antibody titers, and serum insulin levels in high-risk patients .

Unresolved Questions

  • Why only 20–30% of individuals with IAs develop dysglycemia .

  • Role of HLA haplotypes in antibody development .

  • Long-term effects of IgE antibodies on insulin efficacy .

Product Specs

Introduction
Insulin is a hormone that plays a key role in regulating blood sugar levels. It works by increasing the permeability of cells to glucose, allowing them to take up glucose from the bloodstream and use it for energy. Insulin also promotes the storage of glucose as glycogen in the liver and muscles, and it inhibits the production of glucose by the liver. These actions help to keep blood sugar levels within a narrow range.
Description
These antibody-producing cells (clones) were created by fusing myeloma cells (X63-Ag8-653) with spleen cells extracted from Balb/c mice. These mice had been immunized with purified human insulin to trigger an immune response.
Physical Appearance
Clear, sterile solution.
Formulation
This vial contains antibodies dissolved in a phosphate-buffered saline (PBS) solution at a pH of 7.4. To prevent bacterial contamination, the solution also includes 0.09% sodium azide (NaN3) as a preservative.
Stability
For optimal stability, store the antibody solution at a refrigerated temperature of +4 degrees Celsius.
Species Reactivity
While these antibodies primarily target human insulin, they exhibit cross-reactivity with other species' insulin, including: - Human proinsulin - Bovine insulin (30% cross-reactivity) - Porcine insulin Notably, there is no cross-reaction with free C-peptide.
Purification Method

Purified by chromatography on protein A chromatography.

Specificity
Insulin, human.
Type
Mouse Anti Human Monoclonal.
Clone
7F8.
Isotype

IgG1 kappa.

Q&A

What are the different types of insulin antibodies researchers should distinguish between?

Researchers must distinguish between two primary types of insulin antibodies with distinct origins and clinical implications:

Insulin antibodies (IA) are induced by exogenous insulin administration in patients receiving insulin therapy. These develop as an immune response to injected insulin, even when using human insulin formulations. Research indicates that recombinant DNA insulins may be more immunogenic than animal insulins, with studies reporting IA frequencies of 78% to 97% in insulin-treated patients .

Insulin autoantibodies (IAA) occur spontaneously in some patients, predominantly children, prior to type 1 diabetes mellitus (T1DM) diagnosis and before any exposure to exogenous insulin. These autoantibodies are part of the autoimmune process underlying T1DM development .

The distinction is crucial as IA generally exist in much higher concentrations than IAA, leading to different effects on insulin bioavailability and glucose metabolism. Understanding this differentiation is fundamental for accurately interpreting research findings related to insulin antibody phenomena.

How do insulin antibodies affect glycemic control in research subjects?

Anti-insulin antibodies can significantly disrupt glycemic control through several mechanisms that researchers should consider in study design and data interpretation:

Binding-release dynamics: Insulin antibodies act as binding proteins that can sequester circulating insulin, temporarily reducing its bioavailability. This bound insulin can later dissociate from antibodies, creating unpredictable insulin action .

Biphasic dysglycemia pattern: A classic pattern observed is initial post-injection hyperglycemia (due to antibody sequestration of insulin) followed by delayed hypoglycemia (due to unregulated release of bound insulin) .

Clinical presentations in research subjects often include combinations of insulin resistance, nocturnal/matutinal hypoglycemia, and unexplained ketoacidosis . In a study of 40 insulin-treated participants with suspected antibody-mediated dysglycemia, researchers identified 7 subjects with highly likely clinically significant antibodies and 4 with insulin binding of possible clinical significance .

These effects create challenges in research settings, as they introduce variability and unpredictability in insulin pharmacokinetics and pharmacodynamics that may confound study results.

What are the primary indications for testing anti-insulin antibodies in research settings?

Researchers should consider anti-insulin antibody testing in the following research scenarios:

Unexplained glycemic variability: Subjects with insulin-treated diabetes displaying wide and unexplained fluctuations in blood glucose levels despite standardized insulin regimens .

Suspected insulin resistance: Cases requiring unusually high insulin doses that cannot be explained by other factors such as obesity, infection, or medications .

Recurrent unexplained hypoglycemia: Particularly nocturnal or fasting hypoglycemia that occurs despite appropriate insulin dose adjustments .

Apparent allergic responses to insulin: When investigating potential immunologic reactions to insulin administration .

Treatment non-responsiveness: Subjects whose glycemic control does not improve despite escalation of insulin therapy .

Type 1 diabetes risk stratification: When assessing autoimmunity profiles prior to clinical diabetes onset, particularly in pediatric populations or first-degree relatives of T1DM patients .

These indications reflect scenarios where the presence and characteristics of insulin antibodies may provide valuable insights into aberrant glycemic patterns or disease mechanisms. Early detection can inform therapeutic decision-making and potentially identify candidates for immunomodulatory interventions .

How do insulin antibodies differ from other islet autoantibodies in diabetes research?

Insulin autoantibodies (IAA) have distinct characteristics compared to other islet autoantibodies that researchers should account for:

Temporal appearance: IAA often appear earlier in the natural history of T1DM development compared to other islet autoantibodies, particularly in pediatric populations .

Age-dependent significance: IAA have stronger predictive value in younger children, whereas GAD autoantibodies may be more prevalent in adult-onset autoimmune diabetes .

Technical challenges: IAA assays typically have lower sensitivity and specificity compared to other islet autoantibody assays, requiring more standardization and quality control .

Confounding by exogenous insulin: Once insulin therapy begins, distinguishing naturally occurring IAA from treatment-induced IA becomes technically challenging, unlike other islet autoantibodies .

Functional implications: While all islet autoantibodies serve as biomarkers of autoimmunity, insulin antibodies uniquely affect the pharmacokinetics and pharmacodynamics of insulin therapy, directly influencing glycemic control .

A systematic review of 152 studies characterized the heterogeneity in autoantibody profiles, noting that autoantibody type/target was most frequently examined, followed by autoantibody number. Recurring themes included correlations of autoantibody number, type, and titers with progression to clinical diabetes .

How can researchers differentiate between antibody-mediated dysglycemia and other causes of glycemic variability?

Distinguishing antibody-mediated dysglycemia from other causes of glycemic variability requires a systematic approach:

Pattern recognition: Antibody-mediated dysglycemia typically presents with a characteristic pattern of initial hyperglycemia followed by delayed hypoglycemia several hours after insulin administration. Researchers should implement standardized glucose monitoring protocols capturing these temporal patterns .

Laboratory confirmation strategy:

  • Initial screening with total insulin immunoreactivity (IIR)

  • Polyethylene glycol (PEG) precipitation to quantify antibody-bound insulin

  • Gel filtration chromatography (GFC) to confirm high molecular weight insulin immunoreactivity

Quantitative assessment: Research suggests that clinically significant insulin antibodies can be identified using these criteria:

  • IIR above 3000 pmol/L AND

  • 50% reduction in IIR after PEG precipitation AND

  • Substantial high molecular weight IIR on GFC

In a study of 40 participants with suspected antibody-mediated dysglycemia, this systematic approach successfully identified 7 with highly likely clinically significant antibodies that could explain their glycemic lability . This structured methodology allows researchers to accurately identify cases of antibody-mediated dysglycemia for further study or intervention.

What is the prevalence of significant anti-insulin antibodies in insulin-treated diabetes patients?

The prevalence of clinically significant anti-insulin antibodies must be distinguished from the mere presence of detectable antibodies:

Clinically significant antibodies: Based on robust laboratory assessment in a study of 40 insulin-treated participants with suspected antibody-mediated dysglycemia:

  • 7 participants (17.5%) had highly likely clinically significant antibodies

  • 4 participants (10%) had antibodies of possible clinical significance

  • 29 participants (72.5%) had antibodies judged to be insignificant

Stratification by insulin type: Limited data suggests higher immunogenicity with recombinant DNA insulins compared to animal insulins .

These findings highlight the importance of distinguishing between the presence of antibodies and their clinical significance in research settings, with only a minority of insulin-treated patients developing antibodies that significantly affect glycemic control.

How do anti-insulin antibodies manifest differently in pediatric versus adult research cohorts?

Research indicates important age-related differences in insulin antibody manifestations that should inform study design and interpretation:

Insulin autoantibodies (IAA):

  • More prevalent in pediatric-onset T1DM (particularly in children <5 years)

  • Often the first autoantibody to appear in children

  • Higher titers correlate with more rapid progression to clinical diabetes in children

  • Less common as the initial autoantibody in adult-onset autoimmunity

A systematic review of 152 studies found that islet autoantibodies can identify heterogeneity before, at, and after T1D diagnosis, noting that autoantibody number, type, and titers correlate with progression rates. The order of autoantibody seroconversion and interactions with age and genetic background contribute to different phenotypes across age groups .

Clinical manifestations:

  • Different patterns of glycemic variability between pediatric and adult patients

  • Age-specific considerations in laboratory assessment and therapeutic approaches

These differences highlight the importance of age-stratified analysis in insulin antibody research and suggest different pathophysiological mechanisms may predominate across the lifespan.

What are the mechanisms by which insulin antibodies can cause both hypo- and hyperglycemia?

The paradoxical ability of insulin antibodies to cause both hyper- and hypoglycemia stems from complex immunological and pharmacokinetic mechanisms:

Binding-buffering hypothesis:

  • Insulin antibodies act as a buffer system, binding insulin when concentrations are high

  • This initially reduces free insulin availability (causing hyperglycemia)

  • Bound insulin later dissociates unpredictably (causing hypoglycemia)

  • This creates a time-shifted insulin effect disconnected from administration timing

Contributing factors to hyperglycemia:

  • Reduced bioavailability of free insulin

  • Neutralization of insulin's biological activity

  • Accelerated insulin clearance when bound to antibodies

Contributing factors to hypoglycemia:

  • Unregulated release of bound insulin

  • Dissociation occurring during fasting periods

  • Prolonged half-life of antibody-bound insulin complexes

Research has demonstrated that insulin immunoreactivity that falls by more than 50% after PEG precipitation, particularly when above 3000 pmol/L, indicates significant antibody binding that may contribute to clinical dysglycemia . Understanding these mechanisms allows researchers to better interpret glycemic patterns and develop targeted interventions in affected research subjects.

What are the current gold standard laboratory methods for detecting clinically significant insulin antibodies?

A multi-step approach represents the current gold standard for detecting and characterizing clinically significant insulin antibodies:

Initial screening: Broad-specificity insulin immunoassays measuring total insulin immunoreactivity (IIR)

  • Must detect various insulin analogues

  • Should capture antibody-bound insulin

  • Values >3000 pmol/L warrant further investigation

Polyethylene glycol (PEG) precipitation:

  • Separates antibody-bound (precipitated) from free insulin

  • Significant binding defined as >50% reduction in IIR after precipitation

  • Provides quantitative assessment of binding capacity

Gel filtration chromatography (GFC):

  • Confirms high molecular weight insulin immunoreactivity

  • Distinguishes monomeric from antibody-bound insulin

  • Can be enhanced with ex vivo insulin preincubation

This systematic approach has been validated in research settings and allows researchers to distinguish clinically significant insulin antibodies from the frequently occurring but clinically insignificant antibodies in insulin-treated patients. In a study of 40 participants with suspected antibody-mediated dysglycemia, this approach successfully identified those with clinically significant antibodies who might benefit from therapeutic interventions .

How should researchers standardize insulin antibody assays to ensure reproducibility?

Standardization of insulin antibody assays is critical for research reproducibility and requires attention to several key factors:

Participation in standardization programs:

  • Islet Autoantibody Standardization Program

  • Diabetes Antibody Standardization Program

  • Documentation of program participation in methods sections (only 44% of studies explicitly report this)

Sample handling protocol standardization:

  • Consistent collection methods (serum vs. plasma)

  • Standardized processing timeframes

  • Uniform storage conditions

  • Defined freeze-thaw limitations

Assay validation requirements:

  • Intra-assay and inter-assay coefficient of variation documentation

  • Established reference ranges in non-diabetic controls

  • Documented lower limit of detection

Data reporting standards:

  • Reporting of raw values with units

  • Transformation methods clearly described

  • Positivity cut-offs with statistical justification

A systematic review of 152 autoantibody studies demonstrated significant inconsistencies in methodology reporting, with less than half specifically describing participation in standardization programs. Implementing these standardization approaches enhances comparability across studies and increases confidence in reported findings .

What are the technical challenges in differentiating between antibodies to different insulin analogues?

Differentiating between antibodies to different insulin analogues presents several technical challenges that researchers must address:

Structural similarity challenges:

  • High homology between human insulin and analogues

  • Limited epitope differences between analogues

  • Polyclonal antibody responses targeting multiple epitopes

Assay limitations:

  • Commercial assays often lack specificity for different analogues

  • Requirements for custom-developed assays

  • Difficulty standardizing across laboratories

Cross-reactivity assessment:

  • Competitive binding assays required to distinguish specificity

  • Need for specific labeling of different analogues

  • Interpretation of partial vs. complete cross-reactivity

Detection of significant antibodies is particularly complicated by antigenically distinct insulin analogues, necessitating the development of pragmatic biochemical approaches for identifying actionable antibodies . Understanding these challenges and implementing appropriate technical approaches allows researchers to characterize insulin analogue-specific antibodies and their potential clinical implications more accurately.

How can PEG precipitation and gel filtration chromatography be optimized for insulin antibody research?

Optimization of PEG precipitation and gel filtration chromatography (GFC) for insulin antibody research requires attention to multiple methodological details:

PEG precipitation optimization:

  • Calculate percent bound: (Total IIR - Post-PEG IIR)/Total IIR × 100

  • Significant binding threshold: >50% reduction after precipitation

  • Absolute value consideration: Total IIR >3000 pmol/L indicates significance

  • Combined criteria provide better specificity than either alone

GFC protocol refinements:

  • Column selection and buffer composition optimization

  • Ex vivo insulin preincubation to enhance sensitivity

  • Fraction collection and analysis optimization

Combined analytical approach:

  • Initial PEG screening for all samples

  • GFC confirmation for samples with significant PEG precipitation

  • Integration with clinical data for final interpretation

In a study of 40 people with insulin-treated diabetes and suspected antibody-mediated dysglycemia, researchers successfully applied this methodology to categorize participants. Twenty-seven had insulin immunoreactivity (IIR) below 3000 pmol/L with less than 50% reduction after PEG precipitation, indicating insignificant antibodies. Eight had IIR above 3000 pmol/L with more than 50% reduction after PEG precipitation, with GFC confirming substantial high molecular weight IIR in 7 of these 8, indicating likely significant antibodies .

This optimized approach allows more accurate identification of clinically significant insulin antibodies and has been validated in research settings to stratify patients with suspected antibody-mediated dysglycemia.

How can researchers quantify the binding characteristics of insulin antibodies to understand their functional impact?

Comprehensive characterization of insulin antibody binding properties requires sophisticated analytical approaches:

Affinity determination:

  • Radiobinding assays with competitive displacement

  • Advanced analytical techniques for binding kinetics

  • Determination of equilibrium dissociation constants

Capacity quantification:

  • Maximum binding capacity determination

  • Ratio of high to low affinity binding sites

  • Total binding capacity relative to physiological insulin concentrations

  • Correlation with clinical insulin requirements

In vivo correlation strategies:

  • Pharmacokinetic modeling incorporating antibody parameters

  • Continuous glucose monitoring pattern analysis

  • Insulin dose-response assessment

Research has demonstrated that combining immunoassays with PEG precipitation can effectively stratify the clinical significance of insulin antibodies. By examining both the absolute insulin immunoreactivity and the percentage reduction after PEG precipitation, researchers can identify antibodies likely to have functional impacts on insulin action .

These techniques provide insights into how specific binding characteristics (affinity, capacity, kinetics) translate to clinical phenotypes, potentially enabling personalized treatment approaches for patients with significant insulin antibodies.

What are the latest approaches to immunomodulation for treating anti-insulin antibody-mediated dysglycemia?

Research on immunomodulatory approaches for anti-insulin antibody-mediated dysglycemia has yielded several promising strategies:

Immunosuppressive therapy:

  • Successfully implemented in select cases with significant antibodies

  • Monitoring both clinical and biochemical responses is essential

  • Variable clinical outcomes require individualized approaches

Case studies have demonstrated:

  • In two participants with significant antibodies, immunosuppression was initiated

  • One showed good clinical response while another showed only biochemical improvement

  • These varied responses highlight the need for careful patient selection

Conservative management:

  • For six participants, knowledge of underlying antibodies informed adjustment of insulin delivery

  • This approach often proves sufficient for managing less severe cases

  • Understanding antibody characteristics helps optimize insulin regimens

Research indicates that while antibody depletion may be beneficial in severe cases, conservative measures often suffice for managing antibody-mediated dysglycemia in insulin-treated diabetes . This suggests a stepped approach to management, reserving more aggressive immunomodulation for cases that fail conventional optimization of insulin delivery.

How should researchers design studies to evaluate the longitudinal evolution of insulin antibodies?

Designing rigorous longitudinal studies of insulin antibody evolution requires careful consideration of several methodological elements:

Cohort selection and stratification:

  • Include pre-diabetes, new-onset, and established diabetes subjects

  • Stratify by age, genetic risk, and initial antibody status

  • Consider genetic risk scoring for subject classification

Comprehensive antibody profiling:

  • Multiple islet autoantibodies (IAA, GAD, IA-2, ZnT8)

  • Insulin antibody titers, binding capacity, and affinity

  • Epitope specificity and cross-reactivity assessment

A systematic review of 152 studies found that autoantibody features could most strongly be applied to precisely define T1D before diagnosis. The evidence supports continued use of pre-clinical staging paradigms based on autoantibody number and suggests that additional autoantibody features, particularly in relation to age and genetic risk, could offer more precise stratification .

Clinical correlations:

  • Standardized measurement of insulin sensitivity/resistance

  • Continuous glucose monitoring during key study phases

  • C-peptide preservation assessment

  • Insulin dose requirements and hypoglycemia frequency

These design elements enable robust investigation of the dynamic nature of insulin antibody development and its relationship to clinical outcomes.

What are the interactions between genetic factors and insulin antibody development in T1D pathogenesis?

Research into the genetic influences on insulin antibody development reveals complex interactions relevant to T1D pathogenesis:

Genetic risk assessment:

  • Different genetic backgrounds influence autoantibody development patterns

  • Genetic factors interact with age to produce varied autoantibody phenotypes

  • Combined genetic and autoantibody profiles offer improved risk stratification

A systematic review examining relationships between autoantibodies and heterogeneity in T1D found that recurring themes included correlations of autoantibody characteristics with progression rates, differing phenotypes based on the order of autoantibody seroconversion, and interactions with age and genetics .

Translational research applications:

  • Genetic risk scores to identify high-risk subjects for intervention trials

  • Personalized monitoring strategies based on genetic profiles

  • Genetic testing to identify likely responders to immunomodulation

To improve reproducibility and applicability of autoantibody-based precision medicine in T1D, researchers have proposed a methods checklist for islet autoantibody-based manuscripts which includes use of precision medicine MeSH terms and documentation of participation in autoantibody standardization workshops .

Understanding these genetic influences provides insights into the heterogeneity of autoimmune diabetes and may guide personalized approaches to prediction, prevention, and treatment of insulin antibody-mediated phenomena.

Product Science Overview

Introduction

Insulin is a crucial hormone in the regulation of glucose metabolism, produced by the beta cells of the pancreas. It plays a vital role in maintaining blood glucose levels by facilitating the uptake of glucose into cells for energy production and storage. The study of insulin and its interactions is essential for understanding and treating diabetes and other metabolic disorders.

Insulin Structure and Function

Insulin is a peptide hormone composed of 51 amino acids, arranged in two chains (A and B) linked by disulfide bonds. The A chain consists of 21 amino acids, while the B chain has 30 amino acids. Insulin is initially synthesized as a single polypeptide called preproinsulin, which undergoes several processing steps to become mature insulin.

Mouse Anti-Human Insulin Antibodies

Mouse anti-human insulin antibodies are monoclonal antibodies produced by immunizing mice with human insulin. These antibodies are highly specific to human insulin and are used in various research and diagnostic applications. They are particularly valuable in immunohistochemistry (IHC), enzyme-linked immunosorbent assays (ELISA), Western blotting, and flow cytometry.

Applications of Mouse Anti-Human Insulin Antibodies
  1. Immunohistochemistry (IHC): These antibodies are used to detect and visualize insulin in tissue sections. This application is crucial for studying the distribution and localization of insulin-producing cells in the pancreas and other tissues.
  2. Enzyme-Linked Immunosorbent Assay (ELISA): ELISA is a widely used technique for quantifying insulin levels in biological samples. Mouse anti-human insulin antibodies are used as capture or detection antibodies in these assays.
  3. Western Blotting: In Western blotting, these antibodies help detect insulin in protein extracts from cells or tissues. This technique is essential for studying insulin expression and its regulation under various conditions.
  4. Flow Cytometry: Flow cytometry allows for the analysis of insulin-producing cells at the single-cell level. Mouse anti-human insulin antibodies are used to label and quantify these cells in mixed cell populations.
Advantages of Mouse Anti-Human Insulin Antibodies
  • High Specificity: These antibodies are highly specific to human insulin, ensuring accurate detection and quantification.
  • Versatility: They can be used in various applications, including IHC, ELISA, Western blotting, and flow cytometry.
  • Reproducibility: Monoclonal antibodies provide consistent and reproducible results, making them reliable tools for research and diagnostics.

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.