AZGP1 Human

Alpha-2-Glycoprotein 1 Zinc-Binding Human
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Expression

AZGP1 is a 38–40 kDa glycoprotein with structural homology to class I major histocompatibility complex (MHC) proteins but lacks association with β2-microglobulin. Its crystal structure (PDB: 1ZAG) reveals a peptide-binding groove occupied by nonpeptidic ligands, potentially linked to lipid catabolism .

Tissue Distribution:

  • High expression: Secretory epithelial cells (prostate, breast, lung), adipocytes, and bodily fluids (plasma, saliva, urine) .

  • Moderate expression: Liver, stomach, and kidney .

Metabolic Regulation

  • Lipolysis stimulation: AZGP1 induces triglyceride breakdown in adipocytes, contributing to cachexia in advanced cancers .

  • Central nervous system modulation: In hypothalamic POMC neurons, AZGP1 enhances leptin-JAK2-STAT3 signaling, increasing energy expenditure and insulin sensitivity. Overexpression in mice reduces obesity under high-fat diets, while knockout models exhibit metabolic dysfunction .

  • Smoking interaction: Elevated AZGP1 expression due to smoking explains post-cessation weight gain .

Cancer Biomarker

AZGP1’s prognostic value varies by cancer type:

Cancer TypeAZGP1 ExpressionClinical CorrelationSurvival ImpactSource
Gastric cancerLowPoor differentiation, advanced stage5-year survival: 50%
Colon cancerHighTumor progressionReduced DFS/OS
Prostate cancerLowAngiogenesis suppressionSurgical failure risk

Mechanism: In prostate cancer, AZGP1 inhibits angiogenesis by downregulating VEGF .

Disease Biomarkers

  • IgA vasculitis nephritis (IgAVN): Urinary AZGP1 levels are significantly elevated in pediatric patients, showing 85% diagnostic accuracy (AUC: 0.85) .

  • Cardiovascular risk: Elevated serum AZGP1 predicts lower mortality (HR = 0.44) and cardiovascular events (HR = 0.43) in elderly cohorts .

Recombinant Production

Two recombinant variants are widely used:

PropertyE. coli-derived AZGP1 HEK cell-derived AZGP1
Molecular weight34.5 kDa (non-glycosylated)~40 kDa (glycosylated)
BioactivityInduces adipocyte lipolysisBinds fatty acids, regulates melanin
Purity>90% (SDS-PAGE)>90% (RP-HPLC)

Therapeutic Implications

  • Cancer cachexia: AZGP1’s lipid-mobilizing function is leveraged to counteract muscle wasting .

  • Metabolic disorders: Targeting hypothalamic AZGP1 signaling could treat obesity and diabetes .

Product Specs

Introduction
Zinc-alpha-2-glycoprotein (ZAG), also known as tumor-derived lipid mobilizing factor (LMF), is a protein present in various body fluids. It plays a role in lipolysis, body weight regulation, melanin production, and potentially in reducing adiposity. ZAG exhibits a class I major histocompatibility complex (MHC) fold but exists as a soluble protein. It possesses an open apical groove, suggesting the binding of a yet-to-be-identified ligand crucial for its biological activity. Understanding ZAG's interactions with compounds is vital for unraveling its signaling function and potential therapeutic applications.
Description
Human Zinc-Alpha 2 Glycoprotein, derived from Human Serum, has a molecular weight of 32.14 kDa (excluding glycosylation) and comprises 278 amino acids.
Physical Appearance
White, lyophilized powder after filtration.
Formulation
ZA2G protein, after filtration at 0.4 µm, is lyophilized to a concentration of 0.5 mg/ml in a buffer of 20mM TRIS and 50mM NaCl, at pH 8.0.
Solubility
To prepare a working stock solution of about 0.5 mg/ml, add deionized water to the lyophilized pellet and allow it to dissolve completely. This product is not sterile; for cell culture applications, filter it through a sterile filter.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to minimize freeze-thaw cycles. The reconstituted protein remains stable at 4°C for up to two weeks.
Purity
The purity is determined to be greater than 80% using SDS-PAGE analysis.
Human Virus Test
Donor blood samples have been tested and confirmed negative for HBsAg, anti-HCV, HIV Ag/Ab, and syphilis.
Synonyms
Zn-alpha-2-glycoprotein, Zn-alpha-2-GP, AZGP1, ZAG, Zinc-alpha-2-glycoprotein, ZNGP1, ZA2G.
Source
Human Serum.
Amino Acid Sequence
QENQDGRYSL TYIYTGLSKH VEDVPAFQAL GSLNDLQFFR YNSKDRKSQP MGLWRQVEGM EDWKQDSQLQ KAREDIFMET LKDIVEYYND SNGSHVLQGR FGCEIENNRS SGAFWKYYYD GKDYIEFNKE IPAWVPFDPA AQITKQKWEA EPVYVQRAKA YLEEECPATL RKYLKYSKNI LDRQDPPSVV VTSHQAPGEK KKLKCLAYDF YPGKIDVHWT RAGEVQEPEL RGDVLHNGNG TYQSWVVVAV PPQDTAPYSC HVQHSSLAQP LVVPWEAS.

Q&A

What is the chromosomal location and basic structure of the AZGP1 gene?

AZGP1 is located on chromosome 7q22.1 as confirmed by fluorescence in situ hybridization. The gene encodes a soluble protein that functions as a zinc-binding glycoprotein with a molecular weight of 38-40 kDa. Its distinctive electrophoretic mobility within the alpha-2 region contributed to its nomenclature as zinc-alpha-2-glycoprotein (ZAG) . The protein structure shows significant homology with major histocompatibility complex class I molecules, suggesting evolutionary relationships with immune function proteins.

What is the tissue expression profile of AZGP1 and how can it be experimentally determined?

AZGP1 demonstrates broad tissue distribution, being widely expressed in various tissues and body fluids, including the breast, stomach, liver, prostate, plasma, urine, and saliva . Experimental approaches to determine expression include:

  • RT-PCR and Western blot analysis: As demonstrated in colorectal cancer cell studies where AZGP1 expression was assessed across LoVo, HT-29, Caco-2, and HCT116 cell lines

  • Immunohistochemistry: For tissue-specific localization

  • ELISA: For quantification in body fluids

  • RNA-seq: For transcriptomic profiling

Expression ratio analysis comparing target cells to control samples provides quantitative assessment, as seen in the LoVo colorectal cancer cell model where AZGP1 gene/GAPDH expression ratios were measured as 0.35±0.03, compared to 0.78±0.08, 0.67±0.09, and 0.56±0.03 for Caco-2, HT-29, and HCT116 cells, respectively .

How does AZGP1 influence lipid metabolism and body composition?

AZGP1 functions as a lipid-mobilizing factor that stimulates lipolysis and induces body fat reduction in experimental models. Methodologically, this can be investigated through:

  • In vivo studies: Administration of recombinant AZGP1 to mice leads to measurable reduction in body fat

  • Adipocyte culture systems: To evaluate direct effects on lipolysis

  • Metabolomic profiling: To characterize changes in lipid metabolites

Research has established that smoking increases expression of this gene, which explains why smoking cessation often leads to weight gain . This relationship can be experimentally validated through comparative expression studies in smokers versus non-smokers or before and after smoking cessation.

What methodological approaches are recommended for investigating AZGP1's role in diabetes?

Decreased circulating AZGP1 levels are associated with type 2 diabetes . Researchers can investigate this relationship through:

  • Case-control studies: Comparing AZGP1 serum levels between diabetic and non-diabetic subjects

  • Longitudinal cohort studies: Monitoring AZGP1 levels and their correlation with glycemic parameters over time

  • Genetic association studies: Examining AZGP1 polymorphisms in diabetic populations

  • Ex vivo functional assays: Evaluating the effect of varying glucose concentrations on AZGP1 expression in primary tissues

Glucose tolerance tests with concurrent AZGP1 measurement can establish temporal relationships between glycemic fluctuations and protein expression.

How does AZGP1 expression vary across different cancer types and what are the clinical implications?

AZGP1 demonstrates cancer type-specific expression patterns with divergent prognostic implications:

Methodologically, researchers should employ tissue microarrays, immunohistochemistry scoring systems, and survival analysis to establish cancer-specific AZGP1 expression patterns and their correlation with clinical outcomes .

What experimental approaches are most effective for studying AZGP1's functional role in colorectal cancer?

Based on research findings, the following methodological approach has proven effective:

  • Cell line selection: Comparative expression analysis across multiple colorectal cancer cell lines (e.g., LoVo, HT-29, Caco-2, HCT116) to identify appropriate models with differential AZGP1 expression

  • Genetic manipulation: Plasmid-based overexpression systems (e.g., pGCMV/EGFP/AZGP1) with fluorescent markers to confirm transfection efficiency

  • Functional assays:

    • MTT assays for proliferation assessment

    • Colony formation assays (measuring both rate and morphological characteristics)

    • Transwell migration assays to evaluate invasive capacity

In LoVo cells, AZGP1 overexpression resulted in significantly reduced colony formation (4.38%±0.71% vs. 7.15%±0.82% in control) and decreased cell migration (64.33±8.02 vs. 136.67±11.59 migrating cells in control) , demonstrating its tumor-suppressive properties in this context.

Through which signaling pathways does AZGP1 exert its biological effects?

AZGP1 influences cellular function through multiple signaling pathways, prominently including the mTOR pathway. Experimental evidence from LoVo colorectal cancer cells demonstrates that AZGP1 overexpression down-regulates the mTOR signaling pathway components and endogenous fatty acid synthesis . Recommended methodological approaches include:

  • Western blot analysis: To detect changes in pathway components including FASN, eIF4E, p-mTOR, p-S6, and S6K1 proteins

  • RT-PCR: To measure transcript-level changes in pathway components

  • Pathway inhibitor studies: Using specific inhibitors to confirm causal relationships

  • Co-immunoprecipitation: To identify direct protein interactions

Quantitative evaluation reveals that AZGP1 overexpression reduces FASN, eIF4E, and S6K1 gene expression (0.43±0.06, 0.37±0.07, and 0.42±0.04, respectively) compared to control conditions (0.82±0.09, 0.60±0.09, and 0.72±0.06) .

How can researchers effectively study the relationship between AZGP1 and fatty acid metabolism?

AZGP1 regulates endogenous fatty acid synthesis, with implications for both metabolic and cancer research. Methodological approaches include:

  • Gene expression analysis: Measuring FASN and other fatty acid metabolism genes in response to AZGP1 modulation

  • Metabolic labeling: Using isotope-labeled precursors to track fatty acid synthesis rates

  • Lipidomic profiling: Characterizing changes in lipid species composition

  • Fatty acid oxidation assays: Measuring oxidation rates in response to AZGP1 manipulation

Experimental data shows that AZGP1 overexpression in LoVo cells results in significant reduction of FASN expression at both gene and protein levels , suggesting a mechanistic link between AZGP1 and lipid metabolism regulation in cancer cells.

What evidence links AZGP1 to autism spectrum disorders?

Genetic studies have identified AZGP1 variants in autism spectrum disorder (ASD) cases:

  • Two non-synonymous postzygotic mosaic mutations (PZMs) were identified in ASD probands, with statistical significance when compared to background mutation rates (2/571 observed vs. 4/84,448 expected; hypergeometric P-value of 2.7E-04)

  • A de novo frameshift variant in AZGP1 was identified by whole genome sequencing in an ASD proband from a multiplex family

  • AZGP1 has a SFARI Gene Score of 2, indicating moderate evidence for involvement in autism

Researchers investigating this connection should consider:

  • Sequencing strategies that can detect low-level mosaicism

  • Functional characterization of identified variants

  • Expression studies in neural tissues or models

  • Behavioral phenotyping in animal models with AZGP1 mutations

What advanced experimental approaches are recommended for studying AZGP1's role in brain development?

Given the emerging connection between AZGP1 and neurodevelopmental disorders, researchers should consider:

  • Single-cell RNA sequencing: To characterize cell type-specific expression patterns in neural tissues

  • CRISPR/Cas9 genome editing: To model autism-associated mutations in cellular and animal models

  • Patient-derived iPSCs: To generate neural cells carrying AZGP1 variants of interest

  • Spatial transcriptomics: To map AZGP1 expression across brain regions during development

  • Electrophysiological studies: To assess functional consequences of AZGP1 variants on neuronal activity

These methodologies can help elucidate whether AZGP1's effects on neural development are mediated through metabolic pathways, inflammatory signaling, or other mechanisms.

What are the recommended approaches for modulating AZGP1 expression in experimental systems?

Based on published methodologies, researchers can employ several approaches:

  • Plasmid-based overexpression: Using vectors like pGCMV/EGFP/AZGP1 with fluorescent markers for transfection confirmation

  • siRNA or shRNA knockdown: For loss-of-function studies

  • CRISPR/Cas9 gene editing: For generating knockout cell lines or animal models

  • Inducible expression systems: For temporal control of AZGP1 expression

  • Viral vectors: For in vivo delivery to specific tissues

Validation should include both RNA (RT-PCR) and protein (Western blot) level assessment to confirm successful modulation, as demonstrated in the LoVo cell model where overexpression was confirmed by both methods .

How can researchers effectively study protein-protein interactions involving AZGP1?

Understanding AZGP1's interactome is crucial for elucidating its mechanistic functions. Advanced methodological approaches include:

  • Co-immunoprecipitation followed by mass spectrometry: To identify novel binding partners

  • Proximity labeling techniques (BioID, APEX): For capturing transient interactions

  • Yeast two-hybrid screening: For systematic identification of interactors

  • Surface plasmon resonance: For quantitative binding kinetics

  • Fluorescence resonance energy transfer (FRET): For visualizing interactions in living cells

  • Protein fragment complementation assays: For in vivo validation of specific interactions

These approaches can help elucidate how AZGP1 interfaces with the mTOR pathway components and other signaling networks to regulate cellular processes.

How can researchers reconcile the seemingly contradictory roles of AZGP1 across different cancer types?

AZGP1 demonstrates context-dependent functions, acting as a tumor suppressor in some cancers and a negative prognostic factor in others . To address this contradiction, researchers should:

  • Perform comprehensive tissue-specific profiling: Compare AZGP1 expression patterns and signaling effects across multiple cancer types

  • Identify tissue-specific binding partners: Through differential interactome analysis

  • Conduct pathway analysis in multiple contexts: To determine if AZGP1 activates different downstream pathways in different tissues

  • Investigate epigenetic regulation: To determine if different promoter methylation patterns influence function

  • Consider tumor microenvironment: Evaluate how stromal factors might modulate AZGP1's effects

Meta-analysis of expression data across cancer types with correlation to clinical outcomes can provide a comprehensive view of context-dependent functions.

What methodological challenges exist in studying AZGP1's dual role in metabolism and cancer, and how can they be addressed?

AZGP1's involvement in both cancer biology and metabolic regulation presents unique experimental challenges:

  • Cell model selection: Cancer cell lines may not faithfully recapitulate metabolic regulation. Solution: Use primary cells or organoids alongside cancer models

  • Microenvironmental factors: In vitro conditions may not reflect in vivo metabolic milieu. Solution: Employ co-culture systems or conditional media

  • Temporal dynamics: Acute vs. chronic effects may differ. Solution: Use inducible expression systems for temporal control

  • Systemic vs. local effects: Circulating vs. tissue-specific functions may vary. Solution: Combine tissue-specific knockout models with systemic administration studies

  • Cancer-associated cachexia: Separating direct anti-tumor effects from indirect metabolic consequences. Solution: Design experiments that can distinguish cell-autonomous from systemic effects

Integrated multi-omics approaches that simultaneously assess metabolic, transcriptomic, and proteomic changes can provide a more comprehensive understanding of AZGP1's multifaceted functions.

What are the most promising therapeutic applications of AZGP1 research?

Based on current evidence, several therapeutic directions merit investigation:

  • Cancer biomarker development: Further validation of AZGP1 as a prognostic/predictive biomarker, particularly in gastrointestinal and urological cancers

  • Metabolic disease intervention: Exploring recombinant AZGP1 or AZGP1-mimetic compounds for type 2 diabetes and obesity management

  • Cancer cachexia treatment: Targeting AZGP1 pathways to mitigate cancer-associated weight loss

  • mTOR pathway modulation: Leveraging AZGP1's effects on this pathway for cancer therapy

  • Neurodevelopmental disorder therapeutics: Investigating AZGP1-related pathways as potential targets in autism spectrum disorders

Methodologically, researchers should employ translational approaches that bridge basic mechanistic studies with preclinical models and biomarker validation in patient cohorts.

What innovative technologies might advance AZGP1 research in the coming years?

Emerging technologies with potential to transform AZGP1 research include:

  • Spatial transcriptomics: For mapping AZGP1 expression at tissue microenvironmental level

  • Single-cell multi-omics: To correlate AZGP1 expression with proteome, metabolome, and epigenome at single-cell resolution

  • Advanced proteomics: Including hydrogen-deuterium exchange mass spectrometry for detailed protein-protein interaction mapping

  • Cryo-EM and structural biology: For detailed characterization of AZGP1 protein structure and binding interfaces

  • Organoid and microphysiological systems: For studying AZGP1 function in more physiologically relevant contexts

  • AI-driven network analysis: To better understand AZGP1's position in complex biological networks

These technologies can help resolve long-standing questions about AZGP1's multifunctional nature and context-dependent effects across different biological systems.

Product Science Overview

Gene Location and Expression

The AZGP1 gene is located on chromosome 7q22.1 in humans . This gene is widely expressed in various tissues and body fluids, including the breast, stomach, liver, prostate, plasma, urine, and saliva . It is also expressed in secretory cells of lung epithelium .

Biological Functions

AZGP1 plays several important roles in the body:

  • Stimulates Lipolysis: It stimulates lipid degradation in adipocytes, leading to extensive fat loss associated with some advanced cancers .
  • Immune Response: It is involved in the immune response and has been identified as a biomarker in cancer patients .
  • Cell Adhesion and Sensory Perception: It is involved in cell adhesion and the detection of chemical stimuli involved in the sensory perception of bitter taste .
Clinical Significance

AZGP1 has been associated with various diseases and conditions:

  • Cancer: It is known to be a biomarker in cancer patients, with its role varying depending on the specific type of cancer .
  • Diabetes: Decreased circulating levels of AZGP1 are associated with Type 2 diabetes .
  • Weight Regulation: Smoking increases the expression of this gene, which is why smoking cessation often leads to weight gain .
Research and Applications

Research on AZGP1 continues to uncover its various roles and potential applications in medicine. Its ability to stimulate lipolysis and its association with cancer cachexia make it a target for therapeutic interventions .

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.