GLIPR2 Human

GLI Pathogenesis-Related 2 Human Recimbinant
Shipped with Ice Packs
In Stock

Description

Expression Patterns Across Tissues and Cancers

GLIPR2 exhibits tissue-specific expression and dysregulation in malignancies:

Tissue/Cancer TypeExpression ProfileClinical ImplicationSource
LungAbundant expression in normal lung epithelial cells; downregulated in LUADPrognostic biomarker for LUAD progression
ProstateElevated expression in healthy tissue; reduced in prostate adenocarcinomaPotential diagnostic marker
Colorectal/GastricLow expression in colorectal and gastric cancersCorrelated with poor prognosis
Immune-Related TissuesHigh expression in bone marrow, spleen, and peripheral blood leukocytesRole in immune cell regulation

In pan-cancer analyses, GLIPR2 is downregulated in bladder, breast, colon, and pancreatic cancers, while upregulated in cholangiocarcinoma and kidney chromophobe carcinoma .

Tumor Suppression and Therapeutic Sensitivity

GLIPR2 acts as a tumor suppressor by inhibiting migration, invasion, and therapy resistance:

  • In vitro studies: Overexpression in LUAD cell lines (e.g., H1299) reduces migration (Fig. 9C) and invasion (Fig. 9D), while enhancing radiosensitivity (Fig. 9E) and chemosensitivity (Fig. 9F) .

  • Clinical correlation: High GLIPR2 expression in post-radiotherapy LUAD patients predicts better treatment outcomes .

Epithelial-Mesenchymal Transition (EMT)

GLIPR2 promotes EMT in kidney fibrosis through ERK1/2 signaling :

  • Mechanism: Overexpression in HK-2 cells activates ERK1/2, upregulating mesenchymal markers (e.g., vimentin) and enhancing cell migration .

Immune Microenvironment Modulation

GLIPR2 influences immune cell infiltration in tumors:

  • Immune cell recruitment: Enriched in monocytes/macrophages, NK cells, and T cells in single-cell sequencing data .

  • Immunotherapy response: High GLIPR2 expression correlates with favorable clinical outcomes in immunotherapy-treated patients .

Therapeutic and Diagnostic Potential

ApplicationKey FindingsCancer TypeSource
BiomarkerDownregulation predicts LUAD progression; elevated levels correlate with radiosensitivityLUAD
Therapeutic TargetOverexpression enhances treatment sensitivity in vitroNSCLC, CRC
Immunotherapy PredictorHigh expression linked to immune cell infiltration and favorable responsePan-cancer

Mechanistic Insights

GLIPR2 interacts with key cellular pathways:

  1. Autophagy Regulation: Associates with BECN1 (beclin-1) and UVRAG, modulating autophagosome formation .

  2. ERK1/2 Signaling: Activates EMT in kidney fibrosis and cancer progression .

  3. Golgi Function: Localizes to lipid-enriched microdomains, influencing membrane trafficking .

Challenges and Future Directions

  • Translational Limitations: Discrepancies between in vitro and clinical findings (e.g., chemotherapy response in combination therapies) .

  • Research Priorities:

    • Elucidating GLIPR2’s role in hypoxia-driven migration (e.g., HCC) .

    • Investigating its interaction with glycolysis-related genes in CRC .

    • Validating GLIPR2 as a predictive biomarker in larger clinical cohorts .

Product Specs

Introduction
GLIPR2, associated with plant pathogenesis-related (PR-1) proteins, exhibits increased expression in response to pathogen attacks. It resides within lipid-rich microdomains on the cytosolic face of the endomembrane system. Despite lacking a membrane-spanning domain, GLIPR2 strongly associates with membranes and is absent from the cytosol.
Description
Recombinant human GLIPR2, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 174 amino acids (residues 1-154) and possesses a molecular weight of 19.3 kDa. A 20-amino acid His-Tag is fused to the N-terminus of GLIPR2, which undergoes purification using standard chromatographic methods.
Physical Appearance
A clear, colorless solution that has undergone sterile filtration.
Formulation
The GLIPR2 protein solution, at a concentration of 1 mg/ml, consists of 20 mM Tris-HCl (pH 8), 1 mM DTT, 0.1 M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep GLIPR2 refrigerated at 4°C. For extended storage, freeze the protein at -20°C. It is advisable to supplement the protein with a carrier protein like 0.1% HSA or BSA for long-term storage. Repeated freezing and thawing should be avoided.
Purity
SDS-PAGE analysis indicates a purity exceeding 90.0%.
Synonyms
GAPR-1, Golgi-associated plant pathogenesis-related protein 1, GAPR1, GLIPR-2, Golgi-associated PR-1 protein.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGKSASKQFH NEVLKAHNEY RQKHGVPPLK LCKNLNREAQ QYSEALASTR ILKHSPESSR GQCGENLAWA SYDQTGKEVA DRWYSEIKNY NFQQPGFTSG TGHFTAMVWK NTKKMGVGKA SASDGSSFVV ARYFPAGNVV NEGFFEENVL PPKK.

Q&A

What is GLIPR2 and what protein family does it belong to?

GLIPR2 (GLI Pathogenesis Related 2) is a conserved mammalian protein belonging to the pathogenesis related-1 (PR-1) family, containing a sperm-coating protein (SCP) domain of approximately 13 kDa . Unlike other members of the PR-1 family, GLIPR2 is a non-secretory protein because it lacks a signal peptide, making it distinct from other mammalian SCP/CAP domain-containing proteins which are typically secreted . GLIPR2 is primarily expressed in peripheral leukocytes, monocytes, and lung tissue in humans, suggesting a potential role in immunity . The protein can also be referred to by its aliases GAPR-1 and C9orf19 in the scientific literature . Its association with membranes occurs through binding to negatively charged membrane surfaces, a unique characteristic of this protein despite its lack of a conventional signal peptide .

Where is GLIPR2 primarily localized in human cells and what are its basic molecular interactions?

GLIPR2 is primarily a Golgi-associated protein in human cells, where it plays important regulatory roles in cellular processes . Research has shown that GLIPR2 interacts with amino acids 267-284 of BECN1 (Beclin 1), a region that is sufficient to induce autophagy when fused to a cell-penetrating leader sequence . This localization is functionally significant as demonstrated by the observation that GLIPR2 knockout results in less compact Golgi structures, mimicking conditions seen during autophagy induction such as amino acid starvation or Tat-BECN1 peptide treatment . At the molecular level, GLIPR2 has been demonstrated to bind directly to the phosphatidylinositol 3-kinase complex I (PtdIns3K-C1), which consists of PIK3C3/VPS34, PIK3R4/VPS15, BECN1, and ATG14 . This interaction is crucial for understanding GLIPR2's role as a negative regulator of autophagy, as the binding inhibits the lipid kinase activity of PtdIns3K-C1 in vitro .

How does GLIPR2 differ from other PR-1 family proteins?

GLIPR2 exhibits several distinctive characteristics that differentiate it from other pathogenesis related-1 (PR-1) family proteins, making it a unique subject for research . The most notable difference is that while PR-1 proteins and other mammalian SCP domain-containing proteins are secretory proteins, GLIPR2 is a non-secretory protein due to its lack of a signal peptide . This absence of a signal peptide affects its cellular trafficking and localization, resulting in its association with the Golgi apparatus rather than being secreted extracellularly . Another distinguishing feature is GLIPR2's ability to bind directly to negatively charged membranes, which influences its subcellular localization and function . GLIPR2 also has a distinctive expression pattern, being primarily found in peripheral leukocytes, monocytes, and the lung in humans, whereas other PR-1 family proteins may have different tissue distribution patterns . These unique properties of GLIPR2 suggest specialized functions that differ from those of other PR-1 family members, particularly in the context of autophagy regulation and cellular signaling pathways .

What experimental approaches best demonstrate GLIPR2's inhibitory effect on PtdIns3K-C1 activity?

To effectively demonstrate GLIPR2's inhibitory effect on PtdIns3K-C1 activity, researchers should consider multiple complementary experimental approaches that address both in vitro biochemical interactions and cellular consequences . The most direct evidence comes from in vitro lipid kinase assays using purified PtdIns3K-C1 components (PIK3C3/VPS34, PIK3R4/VPS15, BECN1, and ATG14) with and without recombinant GLIPR2 protein, measuring the generation of phosphatidylinositol 3-phosphate (PtdIns3P) . To validate these findings in cellular contexts, researchers should employ CRISPR-Cas9-mediated depletion of GLIPR2 in appropriate cell lines (such as HeLa cells) and measure changes in autophagic flux using LC3-II turnover assays with and without lysosomal inhibitors like bafilomycin A1 or chloroquine . Quantification of PtdIns3P production in cells can be achieved through immunofluorescence detection of WIPI2 puncta formation, as WIPI2 is a PtdIns3P-binding protein whose recruitment serves as a reliable readout for PtdIns3K-C1 activity . Additionally, co-immunoprecipitation experiments should be conducted to confirm the physical interaction between GLIPR2 and components of the PtdIns3K-C1 complex, particularly focusing on the region encompassing amino acids 267-284 of BECN1 .

What evidence supports the role of GLIPR2 in regulating basal autophagy in vivo?

Compelling evidence for GLIPR2's role in regulating basal autophagy in vivo comes from studies using glipr2 knockout mice, which exhibited increased basal autophagic flux across multiple tissues . When designing experiments to further investigate this phenomenon, researchers should employ a comprehensive approach combining tissue-specific analyses of autophagic markers and functional assessments . Immunoblotting for LC3-II and p62/SQSTM1 in tissue lysates from multiple organs of wild-type and glipr2 knockout mice, both under fed conditions and various durations of starvation, would provide a quantitative measure of autophagic flux differences . Immunohistochemical and immunofluorescence analyses of tissue sections should be performed to assess the recruitment of the PtdIns3P-binding protein WIPI2, which serves as a reliable marker for PtdIns3K-C1 activity in vivo . Electron microscopy of tissues from these mice would allow direct visualization and quantification of autophagosome and autolysosome numbers . To establish a causal relationship, tissue-specific rescue experiments using conditional expression of GLIPR2 in knockout backgrounds should be conducted to determine if the enhanced autophagic phenotype can be reversed . Additionally, crossing glipr2 knockout mice with reporter mice expressing GFP-LC3 would facilitate real-time monitoring of autophagy in various tissues and could be particularly valuable for intravital imaging studies of autophagy dynamics in the context of GLIPR2 deficiency .

What are the optimal techniques for studying GLIPR2's interaction with the BECN1-ATG14 complex?

For studying GLIPR2's interaction with the BECN1-ATG14 complex, researchers should employ a multi-faceted approach combining in vitro biochemical assays with cellular and structural biology techniques . Co-immunoprecipitation experiments using either endogenous proteins or tagged versions in relevant cell lines provide the foundation for verifying protein-protein interactions, with specific attention to the region encompassing amino acids 267-284 of BECN1 that has been identified as sufficient for interaction . For more detailed mapping of interaction domains, researchers should conduct in vitro binding assays using purified recombinant proteins or protein fragments, potentially employing techniques such as surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) to determine binding affinities and kinetics . Proximity ligation assays (PLA) can offer visualization of endogenous protein interactions in intact cells with spatial resolution, providing insights into where within the cell these interactions predominantly occur . For structural characterization, X-ray crystallography or cryo-electron microscopy of the GLIPR2-BECN1-ATG14 complex would provide atomic-level details of the interaction interfaces . Additionally, FRET (Förster Resonance Energy Transfer) or BRET (Bioluminescence Resonance Energy Transfer) approaches using fluorescently tagged proteins can reveal dynamic aspects of these interactions in living cells, particularly in response to autophagy-inducing conditions .

What cell culture models and experimental conditions are most appropriate for studying GLIPR2's role in EMT processes?

When investigating GLIPR2's role in epithelial-to-mesenchymal transition (EMT), researchers should carefully select cell culture models and experimental conditions that best recapitulate the physiological context . HK-2 cells (human proximal renal tubular epithelial cells) have been successfully used as an in vitro model system for studying GLIPR2-mediated EMT processes, particularly in the context of kidney fibrosis . For genetic manipulation, stable overexpression systems using vectors like pcDNA3.0-GLIPR2 have proven effective, with G418 selection enabling the establishment of stable cell lines . To comprehensively assess EMT changes, researchers should employ RT-qPCR arrays specifically designed for human EMT processes (such as the SABiosciences RT2 Profiler PCR Array) to identify key affected genes, followed by validation of individual targets . Key EMT markers to monitor include E-cadherin (decreased in EMT), vimentin, and α-SMA (both increased in EMT), using a combination of RT-qPCR, immunoblotting, and immunofluorescence techniques . Functional assays such as cell migration assays (e.g., Transwell migration assays) provide crucial information about EMT-associated phenotypic changes, with quantification of migrating cells serving as an important readout . To investigate signaling pathways involved, particularly the ERK1/2 pathway implicated in GLIPR2-mediated EMT, researchers should use specific inhibitors (e.g., U0126 for MEK/ERK) and siRNA approaches targeting pathway components, monitoring downstream effects on EMT markers and cellular phenotypes .

What are the best approaches for generating and validating GLIPR2 knockout models?

Creating and properly validating GLIPR2 knockout models requires careful consideration of genetic editing strategies, validation methods, and phenotypic characterization approaches . For cellular models, CRISPR-Cas9 technology has proven effective for GLIPR2 depletion, with guide RNAs targeting early exons to ensure complete functional disruption of the protein . When designing guide RNAs, researchers should check for potential off-target effects using prediction tools and select those with minimal off-target potential . Validation of knockout should employ multiple complementary methods including genomic sequencing of the targeted locus to confirm indel formation, Western blotting to verify absence of protein expression, and RT-qPCR to assess transcript levels . For GLIPR2 knockout mice, similar CRISPR-Cas9 approaches can be used, though traditional homologous recombination in embryonic stem cells remains a viable alternative . Phenotypic characterization should be comprehensive, examining multiple tissues and cell types with particular attention to those where GLIPR2 is highly expressed (peripheral leukocytes, lung) or functionally significant (such as kidney tubular cells) . To address potential compensatory mechanisms that may mask phenotypes, researchers should consider acute knockout systems such as inducible Cre-loxP systems or adeno-associated virus (AAV)-delivered Cas9 and guide RNAs for tissue-specific knockout in adult animals . Additionally, rescue experiments reintroducing wild-type or mutant versions of GLIPR2 into knockout backgrounds provide crucial evidence for the specificity of observed phenotypes and can help elucidate structure-function relationships .

How is GLIPR2 expression altered in diabetic nephropathy and what are the implications?

GLIPR2 expression has been found to be significantly elevated in kidney tissue samples from patients with diabetic nephropathy (DN), suggesting its potential involvement in the pathogenesis of this disease . This upregulation appears to be particularly prominent in proximal tubular cells (PTCs), which are key players in the development of renal interstitial fibrosis . The elevated expression of GLIPR2 in diabetic nephropathy has important implications for disease progression, as experimental evidence indicates that GLIPR2 overexpression promotes epithelial-to-mesenchymal transition (EMT) in proximal tubular epithelial cells through activation of the ERK1/2 signaling pathway . This EMT process is characterized by decreased expression of epithelial markers like E-cadherin and increased expression of mesenchymal markers such as vimentin and α-SMA, contributing to the accumulation of interstitial fibroblasts and subsequent fibrosis . Researchers investigating GLIPR2 in diabetic nephropathy should employ immunohistochemical analyses of kidney biopsy samples from DN patients at various disease stages, correlating GLIPR2 expression levels with clinical parameters such as proteinuria, estimated glomerular filtration rate (eGFR), and histopathological indicators of fibrosis . Additionally, studies using diabetic mouse models (such as streptozotocin-induced diabetes or db/db mice) with and without GLIPR2 knockout would help establish whether GLIPR2 inhibition could attenuate diabetic kidney injury and fibrosis .

What is the potential role of GLIPR2 in fibrotic diseases beyond the kidney?

Given GLIPR2's demonstrated role in promoting epithelial-to-mesenchymal transition (EMT) in renal cells and its contribution to kidney fibrosis, there is compelling rationale to investigate its potential involvement in fibrotic diseases affecting other organs . GLIPR2 is expressed in multiple tissues including the lung, where fibrotic conditions like idiopathic pulmonary fibrosis (IPF) share mechanistic similarities with renal fibrosis, particularly regarding the pathological activation of fibroblasts and myofibroblasts . Researchers exploring this question should first perform comparative expression analyses of GLIPR2 across tissue samples from patients with various fibrotic conditions (pulmonary fibrosis, liver cirrhosis, cardiac fibrosis, etc.) versus healthy controls using immunohistochemistry, RT-qPCR, and Western blotting . Cell-type specific localization of GLIPR2 in these tissues should be determined using co-immunofluorescence with markers for specific cell types (epithelial cells, fibroblasts, inflammatory cells) . Functional studies using relevant cell culture models of these tissues (such as lung alveolar epithelial cells, hepatocytes, or cardiac fibroblasts) with GLIPR2 overexpression or knockdown would help establish whether GLIPR2 promotes pro-fibrotic phenotypes through EMT or other mechanisms in these contexts . Additionally, the ERK1/2 pathway should be investigated in these models to determine if GLIPR2's pro-fibrotic effects occur through similar signaling mechanisms across different tissue types .

How might targeting GLIPR2 therapeutically affect autophagy-dependent diseases?

GLIPR2's role as a negative regulator of autophagy makes it a potential therapeutic target for diseases where autophagy dysregulation plays a pathogenic role . When considering therapeutic targeting of GLIPR2, researchers must carefully evaluate both the potential benefits of enhancing autophagy through GLIPR2 inhibition and the possible unintended consequences across different disease contexts . For neurodegenerative diseases characterized by protein aggregation (such as Alzheimer's, Parkinson's, and Huntington's diseases), GLIPR2 inhibition could potentially enhance autophagic clearance of aggregated proteins, as demonstrated by the increased autophagic flux observed in GLIPR2-depleted cells and tissues . Researchers should investigate this possibility using neuronal models expressing disease-associated aggregation-prone proteins (such as tau, α-synuclein, or huntingtin) with GLIPR2 knockdown or overexpression . For certain cancers where autophagy promotes tumor cell survival under stress conditions, inhibiting GLIPR2 might be counterproductive by enhancing this pro-survival mechanism, warranting careful evaluation in cancer models . Methodologically, researchers should develop small molecule inhibitors or peptide-based approaches that disrupt the interaction between GLIPR2 and the BECN1-ATG14 complex, specifically targeting the amino acid region 267-284 of BECN1 that has been identified as critical for this interaction . The efficacy of such approaches should be evaluated by measuring changes in PtdIns3K-C1 activity, PtdIns3P production, and autophagic flux in disease-relevant cell and animal models .

Product Science Overview

Gene and Protein Structure

The GLIPR2 gene is located on chromosome 9 and is a protein-coding gene. The protein encoded by this gene is involved in several critical cellular functions, including protein homodimerization activity, positive regulation of the ERK1 and ERK2 cascade, and epithelial to mesenchymal transition (EMT) . The protein is primarily located in the Golgi membrane, which is essential for its function in cellular processes.

Biological Functions

GLIPR2 plays a significant role in the regulation of epithelial cell migration and EMT. EMT is a process where epithelial cells lose their cell polarity and adhesion properties, gaining migratory and invasive characteristics typical of mesenchymal stem cells. This transition is crucial in various physiological and pathological processes, including embryogenesis, wound healing, and cancer metastasis .

Clinical Significance

GLIPR2 has been studied extensively for its role in cancer. It has been found to be upregulated in several types of cancer, including hepatocellular carcinoma and renal clear cell carcinoma (ccRCC). In ccRCC, GLIPR2 is associated with cellular metastasis and the infiltration of CD8+ T cells . High expression levels of GLIPR2 in ccRCC are correlated with an unfavorable prognosis, making it a potential predictive biomarker for metastasis, immune infiltration, and prognosis .

Research and Therapeutic Potential

Recent studies have highlighted the potential of GLIPR2 as a target for immunotherapeutic interventions. The protein’s involvement in immune cell infiltration and its regulatory role in EMT make it a promising candidate for developing targeted therapies for cancer treatment . Further research is needed to fully understand the mechanisms by which GLIPR2 regulates these processes and to explore its potential in clinical applications.

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.