Recombinant Rat Immediate early response 3-interacting protein 1 (Ier3ip1)

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Rat Immediate Early Response 3-Interacting Protein 1 (Ier3ip1)

Recombinant Rat IER3IP1 (Ier3ip1) is a synthetic version of the endoplasmic reticulum (ER)-resident protein encoded by the IER3IP1 gene. This protein plays critical roles in maintaining ER homeostasis, regulating the unfolded protein response (UPR), and facilitating protein secretion . Recombinant production enables precise functional studies of its interactions, localization, and therapeutic potential, particularly in diseases linked to ER dysfunction, such as diabetes and neurodevelopmental disorders .

Key Features of IER3IP1

PropertyDetails
LocalizationER membrane and Golgi apparatus
Interacting PartnersTMEM167A, Rab11, ERGIC53
FunctionRegulates ER-to-Golgi trafficking, limits UPR activation, supports β-cell survival
Pathogenic MutationsV21G, L78P, T79* (linked to MEDS1: microcephaly, epilepsy, neonatal diabetes)

Functional Insights from Recombinant Studies

Recombinant IER3IP1 is used to:

  1. Map Protein Interactions: Co-immunoprecipitation (Co-IP) and yeast two-hybrid assays confirm binding to TMEM167A and Rab11 .

  2. Study ER Stress: Overexpression or knockout models reveal its role in suppressing XBP1 splicing during UPR activation .

  3. Assess Trafficking Defects: Mutant variants (e.g., V21G) show impaired localization to Rab11 vesicles and reduced interaction with TMEM167A .

Production Parameters

Host SystemPurityTagsApplications
E. coli>85% (SDS-PAGE)His, Avi, FcELISA, Western blotting, Co-IP
Yeast>85%Mammalian tagsStructural studies, secretion assays
Mammalian Cells>85%Native or taggedFunctional assays in β-cells

Key Research Applications

  1. ER Stress Modeling: Recombinant IER3IP1 is used to rescue ER stress in IER3IP1 knockout β-cells and cerebral organoids .

  2. Trafficking Studies: Fluorescence microscopy and biochemical assays track ER-to-Golgi transport defects caused by mutant IER3IP1 .

  3. Disease Modeling: Recombinant proteins enable studying MEDS1-causing mutations in human stem cell-derived islets .

Critical Functions in Cellular Systems

SystemObservationMechanism
B CellsIer3ip1 deficiency causes ER stress, impaired class-switch recombination, and cell cycle arrest .Dysregulated XBP1 signaling and UPR activation.
β-CellsIER3IP1 knockout leads to proinsulin misfolding, reduced insulin secretion, and diabetes .Impaired ER-to-Golgi trafficking of proinsulin.
NeuroblastsInsep (Drosophila homolog) depletion disrupts cytokinesis via Rab11 vesicle fusion defects .IER3IP1 stabilizes Rab11 vesicle docking/fusion.

Disease Relevance

  1. MEDS1 (Microcephaly, Epilepsy, Neonatal Diabetes): Pathogenic IER3IP1 mutations (e.g., V21G, L78P) destabilize the protein, impair ER-to-Golgi trafficking, and trigger UPR-mediated apoptosis .

  2. Type 2 Diabetes: Reduced IER3IP1 expression in β-cells correlates with ER dysfunction and insulin resistance .

Product Specs

Form
Lyophilized powder
Please note: We will prioritize shipping the format currently available in our inventory. If you have specific format requirements, kindly indicate them during order placement. We will accommodate your request to the best of our ability.
Lead Time
Delivery time may vary depending on the purchasing method and location. For specific delivery timelines, please consult your local distributors.
All our proteins are shipped with standard blue ice packs. If dry ice packaging is required, please inform us in advance as additional charges may apply.
Notes
Repeated freezing and thawing is not recommended. For optimal results, store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. To facilitate long-term storage at -20°C/-80°C, we recommend adding 5-50% glycerol (final concentration) and aliquoting the solution. Our standard final glycerol concentration is 50%. Customers can use this as a reference.
Shelf Life
Shelf life is influenced by various factors including storage conditions, buffer composition, temperature, and the protein's intrinsic stability.
Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. Lyophilized form typically has a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. For multiple use, aliquoting is recommended. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
The tag type will be determined during production. If you have a specific tag type in mind, please inform us, and we will prioritize development of your specified tag.
Synonyms
Ier3ip1; Immediate early response 3-interacting protein 1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-82
Protein Length
Full length protein
Species
Rattus norvegicus (Rat)
Target Names
Ier3ip1
Target Protein Sequence
MAFTLYSLMQAALLCVNAIAVLHEERFLKNIGWGTDQGIGGFGEEPGIKSQLLNLIRSVR TVMRVPLIIVNSITIVLLLLFG
Uniprot No.

Target Background

Function
Recombinant Rat Immediate early response 3-interacting protein 1 (Ier3ip1) is a regulator of endoplasmic reticulum secretion that acts as a key determinant of brain size. It is essential for the secretion of extracellular matrix proteins and plays a crucial role in proper brain development by ensuring sufficient deposition of extracellular matrix proteins for tissue integrity and the proliferation of neural progenitors. Additionally, Ier3ip1 functions as a regulator of the unfolded protein response (UPR).
Database Links

UniGene: Rn.7319

Protein Families
YOS1 family
Subcellular Location
Endoplasmic reticulum membrane; Multi-pass membrane protein.

Q&A

What is Ier3ip1 and what are its primary cellular functions?

Ier3ip1 (Immediate early response 3-interacting protein 1) is an endoplasmic reticulum (ER) resident protein highly expressed in pancreatic cells and the developing brain cortex. Its primary function involves facilitating ER-to-Golgi trafficking of proteins, particularly proinsulin in β-cells. Research using CRISPR/Cas9-edited stem cell models has demonstrated that loss of Ier3ip1 results in a threefold reduction in ER-to-Golgi trafficking of proinsulin in stem cell-derived β-cells, leading to cellular dysfunction both in vitro and in vivo . Additionally, Ier3ip1 plays a role in limiting activation of the unfolded protein response (UPR) mediated by inositol-requiring enzyme-1α (IRE1α) and X-box binding protein 1 (XBP1) in B cells, suggesting its importance in maintaining ER homeostasis .

Which animal models are most suitable for studying Ier3ip1 function?

For studying Ier3ip1 function, researchers have successfully employed multiple model systems:

  • Genetically modified mouse models: Forward genetic screening using N-ethyl-N-nitrosourea (ENU)-induced mutations has identified viable hypomorphic Ier3ip1 alleles in mice that faithfully recapitulate aspects of human MEDS (Microcephaly with simplified gyration, Epilepsy, and permanent neonatal Diabetes Syndrome) . These models are particularly valuable for investigating B cell development defects and immune function.

  • Human embryonic stem cell (hESC) models: CRISPR/Cas9-mediated genome editing has been used to generate specific Ier3ip1 mutations in hESCs, which can then be differentiated into pancreatic islet lineages. Two particularly useful models include:

    • Homozygous knock-in of patient mutations (e.g., IER3IP1 V21G)

    • Complete knockout models (IER3IP1−/−)

These models allow for detailed investigation of cell-specific effects of Ier3ip1 deficiency during development and in mature tissues, offering complementary advantages to in vivo studies.

What phenotypes are associated with Ier3ip1 mutations?

Ier3ip1 mutations are associated with several distinct phenotypes across multiple systems:

SystemPhenotypic ManifestationsKey Cellular Mechanisms
PancreaticNeonatal diabetes, reduced insulin secretion, decreased β-cell numberImpaired proinsulin trafficking, elevated ER stress, reduced insulin content (116 vs. 671 ng INS/μg DNA in WT)
NeurologicalMicrocephaly, simplified gyral pattern, epilepsyMistrafficking of proteins crucial for neuronal development including FGFR3, UNC5B, and SEMA4D
ImmunologicalReduced B220+ cells in peripheral blood, B cell development defectsDysregulated unfolded protein response in B cells

The severity of these phenotypes correlates with the nature of the mutation, with complete loss-of-function typically resulting in more severe manifestations than hypomorphic variants.

How does Ier3ip1 facilitate ER-to-Golgi trafficking at the molecular level?

Ier3ip1 facilitates ER-to-Golgi trafficking through multiple coordinated mechanisms:

  • Localization at ER exit sites: Similar to SURF4 (another protein involved in cargo sorting), Ier3ip1 is strategically positioned at ER exit sites, where it contributes to the efficient export of proteins like proinsulin .

  • Cargo receptor interaction: Ier3ip1 forms functional complexes with trafficking mediators including Golgi transmembrane protein 167A . Additionally, the trafficking of cargo receptor ERGIC53 and KDEL-receptor 2 is compromised in the absence of Ier3ip1 .

  • COPII-mediated transport facilitation: While COPII-coated vesicles mediate ER-to-Golgi transport either through receptor-mediated sorting or nonselective bulk flow, Ier3ip1 appears to contribute to the receptor-mediated pathway for specific cargo proteins. The RUSH (Retention Using Selective Hooks) assay has demonstrated that proinsulin accumulation in Ier3ip1-deficient cells is due to defective ER-to-Golgi trafficking .

In β-cells, where proinsulin is the predominant soluble cargo, Ier3ip1's role becomes particularly critical, as evidenced by the 3-fold reduction in proinsulin trafficking observed in Ier3ip1-deficient cells.

What is the relationship between Ier3ip1 deficiency and ER stress?

The relationship between Ier3ip1 deficiency and ER stress involves a complex cascade of molecular events:

  • Upregulation of UPR markers: Ier3ip1-deficient cells show significant upregulation of several ER stress markers, including HSPA5 (BiP), sXBP1, ATF6, and death protein 5 (DP5) . This indicates activation of specific arms of the unfolded protein response.

  • Selective UPR pathway activation: Interestingly, Ier3ip1 deficiency primarily activates the IRE1α and ATF6 arms of the UPR, but not the PERK arm . This selective pathway activation may explain some of the tissue-specific effects observed.

  • Cell-type specific vulnerability: β-cells from Ier3ip1−/− SC-islets show a significantly higher percentage of cells expressing high levels of BiP (31% vs. 7% in wild-type), suggesting particular vulnerability of these cells to ER stress. This is further supported by increased expression of mesencephalic astrocyte-derived neurotrophic factor (MANF) in Ier3ip1−/− β-cells .

  • Differential responses across cell types: While β-cells show increased vulnerability to ER stress-induced apoptosis, α-cells appear more resistant. This may be due to α-cells displaying increased expression of HSPA5 (encoding BiP) and the antiapoptotic gene BCL2L, alongside decreased expression of the proapoptotic gene CHOP in response to Ier3ip1 deficiency .

How do different mutations in Ier3ip1 affect protein function and disease severity?

Different mutations in Ier3ip1 produce varying effects on protein function and disease severity:

Mutation TypeEffect on Protein FunctionPhenotypic SeverityResearch Model
Complete knockout (Ier3ip1−/−)Total loss of functionSevere defects in β-cell number, insulin content (116 vs 671 ng INS/μg DNA), elevated proinsulin:insulin ratio (5 vs 3.5), significant ER stressSC-islet models
Patient mutation (V21G)Partial loss of functionIntermediate phenotype with trend toward decreased insulin (466 ng INS/μg DNA) and increased glucagon content (1.2-fold), minimal ER stress activationSC-islet models
ENU-induced missense mutationsHypomorphic effectsViable animals with severe impairment of B lymphopoiesis, potentially milder neurological/metabolic phenotypesMouse models

These findings suggest a genotype-phenotype correlation where mutation severity directly impacts cellular dysfunction across multiple tissues. The differential effects of mutations on ER stress response may explain some of the variation in clinical presentations, with complete loss-of-function typically associated with more severe MEDS1 phenotypes.

What are the most effective approaches for studying Ier3ip1 trafficking functions?

Several methodological approaches have proven particularly valuable for investigating Ier3ip1's role in protein trafficking:

  • RUSH Assay (Retention Using Selective Hooks): This technique allows for synchronized visualization of cargo trafficking from the ER to the Golgi. In Ier3ip1 studies, it has revealed a threefold reduction in proinsulin trafficking in mutant cells . Implementation requires:

    • Fusion of cargo protein (e.g., proinsulin) with a reporter (typically GFP)

    • Inclusion of a streptavidin-binding peptide for retention

    • Addition of biotin to synchronously release the cargo

    • Time-lapse imaging to track trafficking dynamics

  • High-resolution imaging: Combining confocal microscopy with markers for different cellular compartments provides spatial resolution of trafficking defects. This can be enhanced with:

    • Super-resolution techniques for nanoscale visualization

    • Live-cell imaging for temporal dynamics

    • Quantitative image analysis for measuring colocalization

  • Secretome and cell-surface proteomics: These approaches have successfully identified mistrafficked proteins in Ier3ip1-deficient cells, including those crucial for neuronal development like FGFR3, UNC5B, and SEMA4D . Implementation requires:

    • Cell surface biotinylation or secretome collection

    • Mass spectrometry-based protein identification

    • Quantitative comparison between wild-type and mutant samples

How can researchers optimize CRISPR/Cas9 techniques for generating Ier3ip1 mutant models?

Optimizing CRISPR/Cas9 for generating Ier3ip1 mutant models requires careful attention to several methodological details:

  • Guide RNA design strategy:

    • For knockout models: Target early exons (e.g., exon 1) to maximize disruption

    • For knock-in models: Design guides with cut sites near the desired mutation location

    • Use multiple prediction algorithms to select guides with high on-target and low off-target scores

  • Mutation verification protocols:

    • Perform Sanger sequencing to confirm targeted mutations

    • Verify pluripotency markers expression by qPCR and immunocytochemistry in stem cell models

    • Check for chromosomal abnormalities to ensure genomic integrity

  • Differentiation validation:

    • Assess differentiation markers at each stage (e.g., definitive endoderm, pancreatic progenitors)

    • Quantify cell-type specific populations (e.g., β-cells, α-cells) using flow cytometry

    • Measure functional responses such as insulin secretion under various stimuli

  • Control considerations:

    • Generate multiple mutant clones to account for clonal variation

    • Include isogenic controls whenever possible

    • Consider rescue experiments by reintroducing wild-type Ier3ip1 to confirm phenotype specificity

What assays provide the most sensitive detection of ER-to-Golgi trafficking defects in Ier3ip1 studies?

For sensitive detection of ER-to-Golgi trafficking defects in Ier3ip1 studies, researchers should consider these assays:

  • Dynamic insulin secretion using perifusion assays: This technique measures real-time insulin secretion in response to various stimuli (glucose, GLP-1 analogs, KCl). In Ier3ip1−/− SC-islets, this revealed severely reduced insulin secretion despite preserved stimulus-secretion coupling .

  • Proinsulin:insulin ratio measurement: An elevated ratio (5 vs. 3.5 in wild-type) serves as a sensitive indicator of impaired insulin processing capacity and trafficking defects .

  • ER stress marker quantification: RT-qPCR analysis of stress markers (HSPA5, sXBP1, ATF6, DP5) and immunohistochemical quantification of BiP-positive cells provides indirect evidence of trafficking dysfunction .

  • Subcellular fractionation and biochemical analysis: Isolation of ER, ERGIC, and Golgi fractions followed by immunoblotting for cargo proteins can quantitatively assess trafficking efficiency.

  • Vesicle budding assays: In vitro assays measuring COPII vesicle formation and cargo incorporation from ER membranes can directly assess the mechanistic impact of Ier3ip1 mutations.

How should researchers approach conflicting data on Ier3ip1 function across different cell types?

When confronting conflicting data on Ier3ip1 function across different cell types, researchers should implement a systematic approach:

  • Methodological standardization:

    • Use consistent experimental conditions when comparing across cell types

    • Apply multiple complementary techniques to validate observations

    • Consider the timing of measurements, as temporal dynamics may differ between cell types

  • Contextual analysis:

    • Quantify Ier3ip1 expression levels across cell types being compared

    • Identify cell-type specific interaction partners through techniques like BioID or IP-MS

    • Assess the relative importance of different trafficking pathways in each cell type

  • Integrated data analysis:

    • Develop mathematical models incorporating cell-type specific parameters

    • Use systems biology approaches to identify pathway differences

    • Consider compensatory mechanisms that may be active in specific cell types

  • Reconciliation framework:

    • For example, while Ier3ip1 deficiency affects both β-cells and neurons, the specific cargo proteins affected may differ (proinsulin vs. neuronal development proteins like FGFR3)

    • Similarly, while both β-cells and B cells show Ier3ip1-dependent defects, the relative contribution of ER stress vs. trafficking dysfunction may vary

What are the most challenging aspects of translating Ier3ip1 findings between in vitro and in vivo systems?

Translating Ier3ip1 findings between in vitro and in vivo systems presents several significant challenges:

  • Developmental timing differences:

    • In vivo models reflect cumulative developmental effects of Ier3ip1 deficiency

    • In vitro differentiation protocols may not fully recapitulate developmental transitions

    • Temporal aspects of Ier3ip1 function may be compressed or altered in vitro

  • Microenvironmental factors:

    • In vivo systems include complex cell-cell interactions absent in vitro

    • Hormonal and metabolic influences present in vivo are difficult to replicate

    • For example, SC-islets implanted into mice showed defective human insulin secretion, highlighting the importance of physiological context

  • Species-specific differences:

    • Rat and mouse Ier3ip1 may have subtle functional differences from human IER3IP1

    • Regulatory mechanisms controlling Ier3ip1 expression may vary across species

    • Trafficking pathways may have species-specific components or relative importance

  • Integrated physiological readouts:

    • Connecting molecular phenotypes (trafficking defects) to physiological outcomes

    • Accounting for compensatory mechanisms present in vivo but absent in vitro

    • Distinguishing cell-autonomous from non-cell-autonomous effects

How can researchers differentiate between direct and indirect effects of Ier3ip1 deficiency?

Differentiating between direct and indirect effects of Ier3ip1 deficiency requires careful experimental design:

  • Temporal analysis:

    • Time-course experiments to establish sequential events

    • Inducible knockout/knockdown systems to observe immediate vs. delayed effects

    • Correlation of Ier3ip1 protein levels with phenotypic changes

  • Molecular proximity analysis:

    • Proximity labeling techniques (BioID, APEX) to identify direct interaction partners

    • Co-immunoprecipitation to confirm physical interactions (e.g., with Golgi transmembrane protein 167A)

    • FRET/BRET assays to measure real-time protein-protein interactions

  • Cargo-specific trafficking assays:

    • Direct measurement of specific cargo protein trafficking using RUSH assay

    • Comparison of multiple cargo proteins to identify specificity patterns

    • Structure-function analysis of Ier3ip1 domains involved in cargo recognition

  • Rescue experiments:

    • Selective complementation with Ier3ip1 domains or mutants

    • Introduction of constitutively active downstream effectors

    • Specific inhibition of secondary pathways (e.g., ER stress) to isolate their contribution

How might understanding Ier3ip1 function inform therapeutic approaches for MEDS1?

Understanding Ier3ip1 function opens several potential therapeutic avenues for MEDS1:

  • ER stress modulation:

    • Chemical chaperones (e.g., 4-PBA, TUDCA) could reduce ER stress in Ier3ip1-deficient cells

    • Targeted inhibition of IRE1α or ATF6 pathways might normalize UPR activation

    • The selective upregulation of HSPA5, sXBP1, and ATF6 observed in Ier3ip1−/− SC-islets provides specific targets

  • Trafficking enhancement strategies:

    • Small molecules promoting ER-to-Golgi trafficking could bypass Ier3ip1 deficiency

    • Overexpression of complementary trafficking components like SURF4

    • Peptide-based interventions mimicking Ier3ip1 functional domains

  • Cell replacement approaches:

    • Gene-corrected stem cell-derived β-cells for diabetes management

    • Neuronal progenitor transplantation for neurological manifestations

    • The SC-islet models developed provide proof-of-concept for such approaches

  • Precision-medicine strategies:

    • Mutation-specific interventions for missense variants that might restore partial function

    • Read-through agents for nonsense mutations

    • Splicing modulators for mutations affecting Ier3ip1 mRNA processing

What are the most promising research directions for further elucidating Ier3ip1 mechanisms?

The most promising research directions for further elucidating Ier3ip1 mechanisms include:

  • Structural biology approaches:

    • Cryo-EM or X-ray crystallography of Ier3ip1 alone and in complexes

    • Structure-guided design of functional mimetics

    • Molecular dynamics simulations to understand conformational changes during trafficking

  • Cell-type specific Ier3ip1 interactomes:

    • Comprehensive mapping of Ier3ip1 binding partners across cell types

    • Functional validation of key interactions (e.g., with Golgi transmembrane protein 167A)

    • Temporal dynamics of interaction networks during development and stress

  • Cargo selectivity determinants:

    • Identification of sequence or structural motifs in cargo proteins recognized by Ier3ip1

    • Comprehensive cataloging of Ier3ip1-dependent cargo in multiple cell types

    • Engineering of trafficking pathways with modified cargo selectivity

  • Integrative multi-omics approaches:

    • Combined transcriptomics, proteomics, and metabolomics in Ier3ip1 models

    • Single-cell analyses to capture heterogeneity in response to Ier3ip1 deficiency

    • Computational modeling of Ier3ip1-dependent cellular networks

How can Ier3ip1 research contribute to understanding more common diseases involving ER stress and protein trafficking?

Ier3ip1 research offers valuable insights into more common diseases involving ER stress and protein trafficking:

  • Type 1 and Type 2 diabetes:

    • ER stress is implicated in both forms of diabetes

    • Proinsulin trafficking defects contribute to β-cell dysfunction

    • The mechanistic insights from Ier3ip1 studies on insulin production and secretion could inform therapeutic strategies for more common forms of diabetes

  • Neurodegenerative disorders:

    • Protein trafficking defects and ER stress are hallmarks of conditions like Alzheimer's and Parkinson's

    • Understanding how Ier3ip1 mutations affect neuronal proteins like FGFR3, UNC5B, and SEMA4D could provide insights into more common neurodegeneration mechanisms

  • Autoimmune conditions:

    • The B cell development defects in Ier3ip1 mutant models suggest relevance to immune disorders

    • ER stress in immune cells contributes to autoimmunity

    • Targeting Ier3ip1-related pathways might offer new approaches for modulating immune responses

  • Cancer biology:

    • Altered protein trafficking and ER stress responses are features of many cancers

    • The role of Ier3ip1 in regulating the IRE1α and ATF6 arms of the UPR has implications for cancer cell survival

    • Understanding how cells adapt to trafficking defects could inform cancer therapy development

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 2025 TheBiotek. All Rights Reserved.