IMPAD1 Human, Active

Inositol Monophosphatase Domain Containing 1 Human Recombinant, BioActive
Shipped with Ice Packs
In Stock

Description

IMPAD1 Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 349 amino acids (34-359 a.a.) and having a molecular mass of 37.6kDa.
IMPAD1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
IMPAD1, also known as Inositol Monophosphatase Domain Containing 1, is a protein belonging to the inositol monophosphatase family. Located in the Golgi apparatus, it plays a role in enhancing the hydrolysis of phosphoadenosine phosphate to adenosine monophosphate. Mutations in the IMPAD1 gene can lead to GRAPP type chondrodysplasia, a condition characterized by joint dislocations. Additionally, a pseudogene of IMPAD1 is located on the long arm of chromosome 1.
Description
IMPAD1 Human Recombinant protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It encompasses amino acids 34-359, resulting in a protein of 349 amino acids with a molecular weight of 37.6 kDa. This IMPAD1 protein includes a 23 amino acid His-tag fused to its N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The IMPAD1 solution is provided at a concentration of 0.25 mg/ml in Phosphate-Buffered Saline (pH 7.4).
Stability
For short-term storage (2-4 weeks), the IMPAD1 solution should be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein, such as HSA or BSA (0.1%), is advisable for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of IMPAD1 is determined to be greater than 90.0% by SDS-PAGE analysis.
Biological Activity
The specific activity of IMPAD1 is measured as its ability to dephosphorylate adenosine 3',5'-diphosphate sodium salt at a pH of 7.5 and a temperature of 25°C. It exhibits a specific activity exceeding 3300 pmol/min/ug.
Synonyms

Inositol monophosphatase 3, IMP 3, IMPase 3, EC 3.1.3.25, EC 3.1.3.7, Golgi 3-prime phosphoadenosine 5-prime phosphate 3-prime phosphatase, Golgi-resident PAP phosphatase, gPAPP, Inositol monophosphatase domain-containing protein 1, Inositol-1(or 4)-monophosphatase 3, Myo-inositol monophosphatase A3, IMPAD1, IMPA3, GPAPP, IMP-3

Source

Escherichia Coli.

Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSGRFSLFG LGGEPGGGAA GPAAAADGGT VDLREMLAVS VLAAVRGGDE VRRVRESNVL HEKSKGKTRE GAEDKMTSGD VLSNRKMFYL LKTAFPSVQI NTEEHVDAAD QEVILWDHKI PEDILKEVTT PKEVPAESVT VWIDPLDATQ EYTEDLRKYV TTMVCVAVNG KPMLGVIHKP FSEYTAWAMV DGGSNVKARS SYNEKTPRIV VSRSHSGMVK QVALQTFGNQ TTIIPAGGAG YKVLALLDVP DKSQEKADLY IHVTYIKKWD ICAGNAILKA LGGHMTTLSG EEISYTGSDG IEGGLLASIR MNHQALVRKL PDLEKTGHK

Q&A

What is IMPAD1 and what are its primary biological functions?

IMPAD1 is an inositol monophosphatase domain-containing protein that functions in multiple cellular pathways critical to both normal physiology and disease states . In cancer research, IMPAD1 has been identified as a significant driver of lung cancer invasion and metastasis, with studies showing it promotes invasion by up to 32-fold in experimental models . At the molecular level, IMPAD1 appears to function through at least two major mechanisms: regulation of Golgi-mediated secretion and modulation of mitochondrial metabolism . Specifically, IMPAD1 inhibits mitochondrial Complex I activity, which reduces reactive oxygen species (ROS) levels while simultaneously increasing AMP levels . This metabolic reprogramming activates the AMPK-Notch1-HEY1 signaling pathway, which contributes to cancer progression and metastasis .

How is IMPAD1 expression regulated at the genetic and epigenetic levels?

IMPAD1 has been established as a direct target of epithelial microRNAs, specifically miR-200 and miR~96 . During epithelial-to-mesenchymal transition (EMT), a process critical in cancer progression, these microRNAs are downregulated, resulting in de-repression of IMPAD1 expression . This regulatory mechanism explains how IMPAD1 becomes upregulated during cancer progression, particularly in non-small cell lung cancer (NSCLC) . The connection between EMT and IMPAD1 expression provides insight into how cancer cells acquire invasive and metastatic capabilities through changes in gene expression patterns during transformation .

What experimental models are most appropriate for studying IMPAD1 function?

Studies have successfully employed both in vitro and in vivo models to investigate IMPAD1 function . In vitro, researchers have used NSCLC cell lines including 393P, 344SQ, and human HCC827 with either constitutive or doxycycline-inducible IMPAD1 expression systems . Three-dimensional culture systems utilizing collagen/Matrigel matrices have proven particularly effective for studying IMPAD1's role in invasion, as they better recapitulate the extracellular matrix environment of tumors . For in vivo studies, syngeneic mouse models with subcutaneous implantation of IMPAD1-overexpressing or knockdown cells in 129/Sv mice have successfully demonstrated IMPAD1's role in metastasis while allowing for assessment of tumor microenvironment effects .

How does IMPAD1 mechanistically drive cancer metastasis?

IMPAD1 promotes metastasis through a multi-faceted approach affecting cellular morphology, metabolism, and the secretome . Research demonstrates that IMPAD1 modulates Golgi apparatus morphology and enhances vesicular trafficking through its interaction with Syt11, a trafficking protein . This interaction creates changes in Golgi dynamics that alter both the extracellular matrix composition and the tumor microenvironment (TME) . The IMPAD1-mediated increase in exocytosis facilitates the release of pro-invasive factors, including matrix metalloproteases (MMPs), which can be blocked by pan-MMP inhibitor Ilomastat . Additionally, IMPAD1 creates metabolic changes by inhibiting mitochondrial Complex I, increasing cellular AMP levels, and activating AMPK-Notch1-HEY1 signaling, providing cancer cells with alternative energy pathways under stress conditions .

What is the relationship between IMPAD1 and the AMPK-Notch1-HEY1 signaling pathway?

IMPAD1 activates the AMPK-Notch1-HEY1 signaling pathway through multiple mechanisms . By inhibiting mitochondrial Complex I activity, IMPAD1 triggers a reduction in mitochondrial ROS levels while simultaneously increasing cellular AMP levels . Since AMP acts as an ADORA1 agonist, elevated AMP leads to increased phosphorylation of AMPK (pAMPK) . Research shows that treatment with ADORA1 inhibitor reduces both pAMPK and HEY1 expression in IMPAD1-overexpressing cells, confirming this signaling mechanism . The activated AMPK-Notch1-HEY1 pathway promotes cellular migration, invasion, and ultimately metastasis in lung cancer models . This signaling cascade also provides cancer cells with metabolic flexibility during energy stress conditions, contributing to their survival and aggressive behavior .

How do IMPAD1 and Syt11 interact to regulate vesicular trafficking and metastasis?

IMPAD1 and Syt11 function in an epistatic pathway that critically regulates EMT-mediated vesicular trafficking to drive cancer invasion and metastasis . Mechanistic studies have established that IMPAD1 directly interacts with Syt11, a trafficking protein involved in vesicle fusion and exocytosis . Through this interaction, IMPAD1 promotes a more connected Golgi structure that enhances secretory capacity . Conditioned media from IMPAD1-overexpressing cells significantly increases the invasiveness of non-invasive cancer cells, an effect that can be reversed by Brefeldin A (BFA) treatment, which disrupts Golgi-mediated secretion . The IMPAD1-Syt11 pathway alters the cancer cell secretome composition, modifying the extracellular matrix and tumor microenvironment to create conditions favorable for invasion and metastasis . Importantly, inhibiting either IMPAD1 or Syt11 disrupts this secretory pathway and reverses the invasive phenotype, highlighting the therapeutic potential of targeting this interaction .

What techniques are most effective for assessing IMPAD1-mediated changes in the secretome?

Secretome analysis in IMPAD1 research requires a multi-faceted approach combining functional and analytical techniques . Conditioned media (CM) collection from IMPAD1-overexpressing or knockdown cells, followed by functional invasion assays using non-invasive recipient cells, has proven effective for assessing secretome-mediated effects . Researchers should include Golgi-disrupting agents like Brefeldin A (BFA) as controls to confirm Golgi-dependent secretion mechanisms . For identifying specific secretome components, researchers have successfully used targeted approaches examining known Golgi-secreted proteases such as matrix metalloproteases (MMPs), with validation using specific inhibitors like Ilomastat . Mass spectrometry-based proteomics of CM from IMPAD1-manipulated cells can provide comprehensive secretome profiling, while ELISA and immunoblotting methods offer validation for specific proteins of interest .

What are the recommended approaches for modulating IMPAD1 expression in experimental systems?

For effective IMPAD1 functional studies, researchers have successfully employed several complementary approaches to modulate its expression . For overexpression studies, both constitutive and doxycycline-inducible systems using lentiviral or retroviral vectors have proven effective in multiple cell lines . The inducible approach offers the advantage of temporal control over IMPAD1 expression . For knockdown studies, multiple shRNA approaches targeting different regions of IMPAD1 are recommended to control for off-target effects, with validation of knockdown at both mRNA and protein levels . When conducting in vivo studies, researchers should carefully monitor IMPAD1 expression throughout the experiment, as some knockdown approaches may not maintain suppression over extended periods . For transient modulation, microRNA mimics or inhibitors targeting miR-200 and miR~96 can indirectly regulate IMPAD1 expression by manipulating its natural regulatory mechanisms .

How can researchers effectively evaluate IMPAD1's impact on Golgi morphology and function?

Assessment of IMPAD1's effects on Golgi morphology and function requires integrated microscopic and functional approaches . Immunofluorescence microscopy using Golgi markers (such as GM130, TGN46) in IMPAD1-modulated cells allows visualization of changes in Golgi structure, connectivity, and positioning . Electron microscopy provides higher-resolution analysis of Golgi ultrastructure and vesicle formation . For functional assessment, vesicular trafficking can be monitored using fluorescently tagged cargo proteins or dyes that trace the secretory pathway . Pharmacological interventions with Brefeldin A (BFA), which disrupts the Golgi apparatus, serve as critical controls in these experiments . Researchers should complement these approaches with biochemical fractionation of Golgi membranes followed by proteomic analysis to identify IMPAD1-associated proteins, such as Syt11, that mediate its effects on Golgi function .

How might IMPAD1 be therapeutically targeted in cancer treatment?

Given IMPAD1's role in promoting cancer metastasis through multiple mechanisms, several potential therapeutic approaches could be developed . Direct inhibition of IMPAD1 enzymatic activity could be achieved through small molecule inhibitors designed to target its active site, though this requires detailed structural studies of the protein . Alternatively, disrupting the IMPAD1-Syt11 interaction using peptide mimetics or small molecules could inhibit its effects on Golgi trafficking . Upstream regulation offers another approach, where delivery of miR-200 or miR~96 mimics could suppress IMPAD1 expression . Targeting downstream effectors presents additional options, such as inhibiting matrix metalloproteases using agents like Ilomastat, or disrupting the AMPK-Notch1-HEY1 signaling with pathway-specific inhibitors . For potential therapeutic development, researchers should first validate these approaches in patient-derived xenograft models to assess efficacy and toxicity profiles before clinical translation .

What connections exist between IMPAD1 and other therapeutic targets in cancer?

Research suggests potential interactions between IMPAD1 and established therapeutic targets that warrant further investigation . Intriguingly, studies have identified a possible connection between lithium treatment and cancer pathways involving IMPAD1 . Analyses of lithium-sensitive interactomes showed mutual enrichment between IMPAD1 and various cancer-related signaling pathways, suggesting lithium might modulate IMPAD1-dependent oncogenic mechanisms . Additionally, the AMPK-Notch1-HEY1 pathway activated by IMPAD1 overlaps with several targetable oncogenic pathways . The connection to ADORA1 (adenosine A1 receptor) is particularly relevant, as ADORA1 inhibitors reduced pAMPK and HEY1 expression in IMPAD1-overexpressing cells, suggesting adenosine receptor antagonists might counteract IMPAD1-mediated effects . For comprehensive cancer therapy development, researchers should investigate combination approaches targeting both IMPAD1 and these connected pathways to potentially achieve synergistic anti-metastatic effects .

What are the critical knowledge gaps in IMPAD1 research?

Despite significant advances in understanding IMPAD1's role in cancer, several knowledge gaps remain that warrant investigation . The precise enzymatic activities of IMPAD1 in its active human form need further biochemical characterization, particularly regarding substrate specificity and reaction kinetics . While IMPAD1's role in cancer is increasingly clear, its functions in normal physiology remain largely undefined . The complete composition of the IMPAD1-influenced secretome requires comprehensive proteomic characterization to identify all factors contributing to its pro-metastatic effects . Additionally, the detailed structural basis of the IMPAD1-Syt11 interaction needs crystallographic or cryo-EM studies to facilitate targeted disruption approaches . Research into potential IMPAD1 polymorphisms and mutations across different patient populations would help understand its variable expression and activity in cancer subtypes . Finally, integration of IMPAD1 function with broader cellular stress responses and metabolic adaptation during metastasis represents an important area for further exploration .

How might emerging technologies advance IMPAD1 research?

Emerging technologies offer promising approaches to address existing challenges in IMPAD1 research . CRISPR-Cas9 genetic screens focusing on synthetic lethality could identify critical dependencies in IMPAD1-overexpressing cancer cells, potentially revealing new therapeutic vulnerabilities . Single-cell transcriptomics and proteomics would help characterize heterogeneity in IMPAD1 expression within tumors and identify specific cell populations driving metastasis . Spatial transcriptomics and proteomics technologies could map IMPAD1 expression patterns in relation to tumor microenvironment components, providing insight into its role in cancer-stroma interactions . Advanced imaging techniques such as super-resolution microscopy and live-cell imaging could better visualize IMPAD1's effects on Golgi dynamics and vesicular trafficking in real-time . Computational approaches integrating multi-omics data could model IMPAD1 regulatory networks and predict optimal combination therapy strategies . Finally, patient-derived organoids and humanized mouse models would provide more physiologically relevant systems for testing IMPAD1-targeted interventions before clinical translation .

Product Science Overview

Structure and Function

IMPAD1 is involved in the hydrolysis of inositol monophosphate to produce free inositol, which is a key component in the synthesis of phosphatidylinositol and its derivatives. These molecules are essential for various cellular processes, including signal transduction, membrane trafficking, and cytoskeletal organization.

Recombinant Production

Recombinant Human Inositol Monophosphatase Domain Containing 1 (BioActive) is produced using recombinant DNA technology. This involves inserting the human IMPAD1 gene into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. The recombinant protein is then purified and tested for bioactivity to ensure it retains its functional properties.

Applications

The bioactive recombinant IMPAD1 protein is used in various research applications, including:

  • Biochemical studies: Understanding the enzyme’s role in cellular processes and its regulation.
  • Drug discovery: Screening for potential inhibitors or activators of IMPAD1, which could be therapeutic targets for diseases related to phosphatidylinositol signaling.
  • Structural biology: Investigating the three-dimensional structure of IMPAD1 to gain insights into its mechanism of action.
Clinical Relevance

Mutations in the IMPAD1 gene have been associated with certain medical conditions, such as chondrodysplasia with joint dislocations, GPAPP type. This highlights the importance of IMPAD1 in normal skeletal development and its potential as a target for therapeutic intervention.

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.