LRG1 Human, Sf9

Leucine-Rich Alpha-2-Glycoprotein 1 Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

Production and Purification

LRG1 Human, Sf9 is expressed via baculovirus vectors and purified using immobilized metal affinity chromatography (IMAC) targeting the His tag. Critical steps include:

  • Expression: Optimized in Sf9 cells for high yield (~0.5 mg/mL) .

  • Purification: Proprietary chromatographic techniques ensure minimal endotoxin contamination .

  • Quality Control: Validated by SDS-PAGE and HPLC for consistency .

Functional Insights

LRG1 plays multifaceted roles in physiology and disease, with Sf9-derived LRG1 used to study:

Angiogenesis and TGF-β Signaling

  • LRG1 binds endoglin and activates the ALK1-Smad1/5/8 pathway, promoting pathological angiogenesis in cancers and eye diseases .

  • In vitro, LRG1 Human, Sf9 induces endothelial cell sprouting and tube formation, mimicking tumor microenvironment effects .

Metabolic Regulation

  • LRG1 acts as an adipokine, enhancing insulin sensitivity in obese mice. Overexpression reduces fasting glucose and HOMA-IR scores by ~50% .

  • Serum LRG1 levels correlate with obesity and inflammation, suggesting a dual role in metabolic syndrome .

Cancer Progression

  • LRG1 promotes colorectal cancer growth via HER3 activation (KD = 100 nM), independent of TGF-β signaling .

  • Neutralizing antibodies against LRG1 (e.g., 15C4) inhibit tumor growth in murine models, extending survival by 40% .

Research Applications

LRG1 Human, Sf9 is utilized in:

  • Mechanistic Studies: Investigating TGF-β and HER3 signaling pathways .

  • Therapeutic Development: Screening anti-angiogenic or metabolic drugs .

  • Biomarker Research: Detecting elevated LRG1 in pancreatic cancer (AUC = 0.89 in plasma ELISA) .

Table 2: Comparative Analysis of LRG1 Production Systems

ParameterSf9 Baculovirus E. coli
GlycosylationYesNo
Molecular Weight35.4 kDa36 kDa
TagC-terminal 6xHisN-terminal 6xHis
YieldHigh (~0.5 mg/mL)Moderate
ApplicationsFunctional assays, therapyStructural studies, ELISA

Product Specs

Introduction

LRG1, a member of the leucine-rich repeat (LRR) protein family, plays a crucial role in various cellular processes, including protein-protein interactions, signal transduction, cell adhesion, and development. Notably, LRG1 expression is observed during the differentiation of granulocytes.

Description

Recombinant LRG1, expressed in Sf9 insect cells using a baculovirus system, is a single, glycosylated polypeptide chain consisting of 321 amino acids (residues 36-347). With a molecular weight of 35.4 kDa, this protein is engineered with a 6-amino acid Histidine tag at the C-terminus to facilitate purification via proprietary chromatographic techniques.

Physical Appearance
Sterile Filtered colorless solution.
Formulation

The LRG1 protein solution is provided at a concentration of 0.5 mg/ml and is formulated in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.

Stability
For short-term storage (2-4 weeks), the LRG1 protein solution can be stored at 4°C. For extended storage, it is recommended to store the protein at -20°C. To ensure optimal stability during long-term storage, the addition of a carrier protein (0.1% HSA or BSA) is advised. Avoid repeated freeze-thaw cycles to maintain protein integrity.
Purity

The purity of the LRG1 protein is determined to be greater than 95.0% by SDS-PAGE analysis.

Synonyms

Leucine Rich Alpha-2-Glycoprotein 1, Leucine-Rich Alpha-2-Glycoprotein, 1300008B03Rik, 2310031E04Rik, HMFT1766, Leucine-rich alpha-2-glycoprotein, LRG1, LRG. 

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ADPVTLSPKD CQVFRSDHGS SISCQPPAEI PGYLPADTVH LAVEFFNLTH LPANLLQGAS KLQELHLSSN GLESLSPEFL RPVPQLRVLD LTRNALTGLP PGLFQASATL DTLVLKENQL EVLEVSWLHG LKALGHLDLS GNRLRKLPPG LLANFTLLRT LDLGENQLET LPPDLLRGPL QLERLHLEGN KLQVLGKDLL LPQPDLRYLF LNGNKLARVA AGAFQGLRQL DMLDLSNNSL ASVPEGLWAS LGQPNWDMRD GFDISGNPWI CDQNLSDLYR WLQAQKDKMF SQNDTRCAGP EAVKGQTLLA VAKSQHHHHH H

Q&A

What is the molecular structure of human LRG1 and how does it relate to its function?

LRG1 is a secreted glycoprotein with a distinctive horseshoe-like solenoid structure characteristic of leucine-rich repeat proteins. The crystal structure analysis reveals four N-glycosylation sites that significantly impact its biological activity . The protein's horseshoe shape facilitates its interaction with various signaling molecules, particularly in the TGFβ pathway. This structural arrangement allows LRG1 to function as a versatile signaling modulator across multiple physiological and pathological processes .

How do the signaling pathways modulated by LRG1 contribute to disease pathogenesis?

LRG1 primarily modulates the TGFβ signaling pathway, acting as an upstream modifier in a context-dependent manner . Recent research has also identified LPHN2 as a TGF-β-independent receptor for LRG1 . Through these pathways, LRG1 influences multiple cellular processes including angiogenesis, fibrosis, inflammation, and immune cell function. In pathological conditions, LRG1 can disrupt normal cellular interactions required for vascular maintenance, leading to aberrant neovascularization and contributing to the establishment of hypoxic and immunosuppressive microenvironments . Additionally, LRG1 affects epithelial, immune, mesenchymal, and cancer cells through its modulation of TGFβ signaling, directly contributing to disease progression in various contexts.

How can researchers optimize LRG1 purification protocols to maintain protein functionality?

Optimizing LRG1 purification requires careful consideration of several factors. Initial clarification steps should minimize proteolytic degradation by including appropriate protease inhibitors. Affinity chromatography using engineered tags (His, FLAG, etc.) provides efficient capture while maintaining native conformation. Since LRG1 is a glycoprotein, purification conditions must preserve its glycosylation profile, avoiding harsh elution conditions. Size exclusion chromatography as a polishing step helps ensure removal of aggregates and degradation products. Throughout purification, protein functionality should be monitored using activity assays that assess TGFβ pathway modulation or LPHN2 binding . Glycan integrity can be verified using lectin blotting or mass spectrometry. Storage conditions (-80°C in buffers containing glycerol or lyophilized) should be validated to ensure long-term stability and functionality.

What analytical methods are most effective for characterizing recombinant LRG1 quality and identity?

Comprehensive characterization of recombinant LRG1 requires multiple complementary analytical methods. SDS-PAGE and western blotting confirm protein size and identity, while mass spectrometry provides precise molecular weight determination and sequence verification . For glycosylation analysis, specialized techniques such as glycan profiling by HPLC or mass spectrometry are essential to characterize the four N-glycosylation sites identified in the crystal structure . Circular dichroism spectroscopy assesses proper protein folding and secondary structure content. Functional characterization through cell-based assays measuring TGFβ pathway modulation or LPHN2 receptor binding confirms biological activity. Finally, stability studies under various storage conditions ensure the protein maintains its structural and functional integrity over time.

How can researchers establish reliable cellular assays to measure LRG1's effects on TGFβ signaling?

Establishing reliable cellular assays for LRG1's effects on TGFβ signaling requires a multi-faceted approach. Researchers should first select appropriate cell lines that express relevant TGFβ receptors (particularly endothelial cells, fibroblasts, or immune cells). Reporter assays using cells transfected with SMAD-responsive luciferase constructs provide quantitative readouts of canonical TGFβ signaling. Western blotting for phosphorylated SMAD proteins (both SMAD2/3 and SMAD1/5/8) can distinguish between different TGFβ signaling branches . To isolate LRG1-specific effects, parallel experiments with TGFβ receptor inhibitors, neutralizing antibodies, or receptor knockdowns are essential. Time-course experiments are particularly important as LRG1 may show different effects at early versus late timepoints. Finally, downstream functional readouts such as gene expression changes, cell proliferation, migration, or extracellular matrix production provide a more comprehensive view of LRG1's impact on TGFβ signaling.

What experimental approaches help distinguish LRG1's direct effects from secondary responses in complex disease models?

Distinguishing LRG1's direct effects from secondary responses in complex disease models requires sophisticated experimental designs. Conditional knockout or knockdown models with tissue-specific or temporal control allow researchers to determine cell-autonomous versus non-autonomous effects. Cell type-specific reporter systems tracking LRG1 expression and activity in vivo provide spatial and temporal resolution. Ex vivo approaches using isolated tissues or cells from disease models treated with recombinant LRG1 or neutralizing antibodies can isolate direct responses. Single-cell RNA sequencing helps identify primary responder cells versus secondary effectors. Importantly, comparing acute versus chronic LRG1 manipulation can separate immediate signaling effects from adaptive responses . Careful kinetic studies following LRG1 administration or inhibition are essential, as studies in acute calculus cholecystitis showed LRG1's correlation with symptom duration was significantly better than conventional markers .

How does glycosylation pattern variation in LRG1 affect its functional properties and experimental outcomes?

Glycosylation pattern variation significantly impacts LRG1's functional properties and experimental outcomes. The crystal structure of LRG1 revealed four N-glycosylation sites, with specific sites being crucial for receptor interactions . Research shows that serum LRG1 and neutrophil-derived LRG1 have different molecular weights due to differential glycosylation, which affects their biological functions . When using Sf9-expressed LRG1, researchers must account for the insect cell-specific glycosylation patterns, which lack complex mammalian-type glycans and terminal sialylation. This limitation can be addressed through glycoengineering approaches or comparison with mammalian cell-expressed LRG1. Experimental designs should include glycosylation analysis to correlate structural variations with functional outcomes. Particularly notable is that de-glycosylation at one specific site appears critical for LRG1's interaction with the LPHN2 receptor, which mediates vascular and neurological functions in diabetic conditions .

How do researchers quantitatively analyze LRG1 as a biomarker in clinical samples?

Quantitative analysis of LRG1 as a biomarker in clinical samples employs several validated methodologies. ELISA represents the most widely used approach, with commercially available human LRG1 kits demonstrating high sensitivity and specificity . For instance, in acute cholecystitis studies, researchers used ELISA kits with a detection range of 3-96 μg/mL, finding a cut-off value of 6.46 μg/mL with 60% sensitivity and 63% specificity . Mass spectrometry provides an alternative approach with the advantage of detecting specific isoforms and post-translational modifications, as employed in longitudinal studies of hypertensive patients at risk for HFpEF . Western blotting quantifies LRG1 in tissue samples, as demonstrated in left ventricular tissue analysis from HFpEF patients versus controls . For standardization, researchers should include appropriate reference standards, validate assay performance characteristics (precision, accuracy, linearity), and account for potential confounding factors such as sample collection methods, storage conditions, and patient demographics.

What experimental models best recapitulate LRG1's role in cardiac pathophysiology and fibrosis?

The optimal experimental models for studying LRG1's role in cardiac pathophysiology and fibrosis span in vitro, ex vivo, and in vivo approaches. In vitro, primary human cardiac fibroblasts stimulated with TGF-β and subjected to LRG1 modulation have proven valuable for investigating fibroblast activation and fibrosis . These systems allow precise manipulation of LRG1 levels to elucidate its role in fibroblast function. For tissue-level studies, myocardial tissue slices and engineered heart tissues provide intermediate complexity while maintaining tissue architecture. In vivo models include pressure overload (transverse aortic constriction), volume overload (aortic regurgitation), and hypertension models that develop features of HFpEF. Importantly, longitudinal studies in asymptomatic hypertensive patients at risk of developing HFpEF have revealed correlations between circulating LRG1 levels, BNP, and left atrial volume index (LAVI) . Such translational approaches bridge animal models and human disease, with left ventricular tissue samples from HFpEF patients showing significantly elevated LRG1 expression compared to controls .

How can researchers effectively design experiments to evaluate LRG1's dual roles in inflammation and tissue repair?

Designing experiments to evaluate LRG1's dual roles requires sophisticated approaches that capture both temporal dynamics and microenvironmental context. Time-course studies are essential, as LRG1 may promote initial inflammatory responses while later facilitating resolution and repair. For instance, in acute cholecystitis, LRG1 showed superior correlation with symptom duration compared to traditional markers like CRP and WBC . Cell-type specific conditional knockout models help dissect LRG1's effects on different cellular players in inflammation and repair. Co-culture systems incorporating immune cells, fibroblasts, and parenchymal cells allow study of intercellular communication. Varying experimental conditions to mimic different disease phases is crucial—acute high-dose inflammatory stimuli versus chronic low-grade inflammation produce distinct LRG1 responses. Importantly, employing Lrg1-/- mouse models has shown that while LRG1 appears redundant in development and homeostasis, it plays significant roles in pathological conditions . Functional readouts should assess both inflammatory parameters (cytokine production, immune cell recruitment) and repair processes (angiogenesis, extracellular matrix remodeling, tissue regeneration).

What are the cutting-edge approaches for mapping LRG1's interactome in different cellular contexts?

Cutting-edge approaches for mapping LRG1's interactome include proximity-based labeling techniques like BioID or APEX2, where LRG1 is fused to a promiscuous biotin ligase that tags nearby proteins for subsequent identification by mass spectrometry. This approach captures even transient interactions in living cells. Cross-linking mass spectrometry (XL-MS) covalently links interacting proteins before analysis, providing structural insights into interaction interfaces. For receptor interactions, particularly with TGFβ receptors and LPHN2, surface plasmon resonance and bio-layer interferometry measure binding kinetics and affinities with purified components . Cell-specific interactome analysis using FACS-sorted cells from transgenic models expressing tagged LRG1 provides physiologically relevant interaction maps. Computational approaches integrating structural data from the resolved crystal structure with molecular docking can predict novel interactions . These techniques have already revealed LRG1's interactions with key signaling molecules and receptors that mediate its diverse functions in different cellular contexts.

How can researchers resolve contradictory findings regarding LRG1's role across different disease models?

Resolving contradictory findings regarding LRG1 requires systematic investigation of context-dependent factors. Researchers should directly compare multiple disease models under standardized conditions, controlling for variables such as genetic background, age, sex, and environmental factors. Detailed characterization of LRG1 expression patterns, including cell type-specific sources and temporal dynamics, is essential as studies show LRG1 can be secreted from various cell types in different tissue microenvironments . Dose-response relationships should be carefully established, as LRG1 may exhibit hormetic effects—beneficial at certain concentrations but detrimental at others. The protein's post-translational modifications, particularly glycosylation patterns, must be characterized as they significantly impact function . Mechanistic studies should elucidate pathway-specific effects, as LRG1 modulates multiple signaling cascades in a context-dependent manner . Finally, translational validation in human samples helps determine which experimental models best recapitulate human pathophysiology, as demonstrated in studies correlating LRG1 levels with disease progression in HFpEF patients .

What are the methodological considerations for developing and testing therapeutic approaches targeting LRG1?

Developing and testing therapeutic approaches targeting LRG1 requires careful methodological considerations. For function-blocking antibodies, epitope mapping based on structural data helps identify neutralizing regions without affecting beneficial functions. Multiple blocking strategies should be tested, including direct LRG1 neutralization, receptor antagonism, and signaling pathway inhibition. Timing of intervention is critical—therapeutic windows may differ across diseases, as suggested by LRG1's dynamic expression in acute versus chronic conditions . Delivery methods must ensure target engagement in relevant tissues, which may require tissue-specific targeting strategies. Biomarker development is essential for patient stratification and monitoring treatment response, with evidence showing LRG1 correlates with disease parameters like LAVI in cardiac remodeling . Safety assessments must consider potential on-target adverse effects, particularly on beneficial LRG1 functions in tissue repair and innate immunity . Combination approaches with standard-of-care treatments should be evaluated to assess additive or synergistic effects. Importantly, animal studies have demonstrated that LRG1 inhibition through gene deletion or function-blocking antibodies can attenuate disease progression, providing strong preclinical rationale for therapeutic development .

How might single-cell technologies advance our understanding of cell-specific LRG1 responses?

Single-cell technologies offer unprecedented opportunities to dissect cell-specific LRG1 responses across diverse physiological and pathological contexts. Single-cell RNA sequencing can identify which cell populations produce LRG1 and which respond to it, revealing previously unrecognized cellular sources beyond the known hepatocytes and neutrophils . Single-cell proteomics and phosphoproteomics can map signaling cascades activated by LRG1 in individual cells, uncovering heterogeneous responses within seemingly uniform populations. Spatial transcriptomics and proteomics maintain tissue architecture information, enabling correlation of LRG1 expression patterns with microenvironmental features and neighboring cell interactions. Single-cell ATAC-seq reveals how LRG1 signaling affects chromatin accessibility and epigenetic regulation in different cell types. These technologies can be applied to complex disease tissues, such as cardiac samples from HFpEF patients, where LRG1 shows elevated expression , potentially uncovering novel cellular targets and mechanisms that traditional bulk analyses would miss.

What strategies can overcome the challenges of studying glycosylation heterogeneity in LRG1 biology?

Addressing glycosylation heterogeneity in LRG1 biology requires integrated technological and experimental approaches. Mass spectrometry-based glycoproteomics can characterize site-specific glycan structures at LRG1's four N-glycosylation sites identified in its crystal structure . Glycoengineering in expression systems, including modified Sf9 cells with humanized glycosylation pathways, produces more homogeneous glycoforms for functional studies. CRISPR-based glycosylation site mutagenesis creates defined glycovariants to assess structure-function relationships. Lectin arrays and glycan-specific antibodies enable rapid profiling of glycan patterns across different sources of LRG1. Native mass spectrometry preserves intact glycoprotein analysis, revealing the distribution of glycoforms. Importantly, comparative studies between recombinant and native LRG1 from various sources (serum, neutrophils, tissue) help establish the functional significance of glycan differences . These approaches are essential since LRG1 from different cellular sources shows varying molecular weights and biological activities due to differential glycosylation.

How can computational modeling integrate structural, functional, and clinical data to guide LRG1 research?

Computational modeling provides powerful frameworks for integrating multi-dimensional LRG1 data. Molecular dynamics simulations based on the crystal structure of LRG1 can predict how glycosylation or mutations affect protein conformation and receptor interactions . Network analysis integrating proteomics, transcriptomics, and interactome data creates comprehensive maps of LRG1-regulated pathways across different cellular contexts. Machine learning approaches applied to biomarker data can identify patient subgroups most likely to benefit from LRG1-targeted therapies, particularly important in heterogeneous conditions like HFpEF where LRG1 levels correlate with disease progression markers . Systems biology modeling incorporating feedback loops and temporal dynamics helps predict LRG1's context-dependent effects in complex disease processes. Pharmacokinetic/pharmacodynamic modeling guides therapeutic development by predicting optimal dosing regimens and treatment schedules. These computational approaches generate testable hypotheses for experimental validation and accelerate translational applications by prioritizing the most promising research directions based on integrated evidence across structural, mechanistic, and clinical domains.

Product Science Overview

Structure and Expression

LRG1 is a secreted glycoprotein that is abundantly present in the microenvironment of many tumors, where it contributes to vascular dysfunction . The recombinant form of LRG1, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain containing 321 amino acids and has a molecular mass of 35.4 kDa . This recombinant protein is expressed with a 6-amino acid His tag at the C-terminus and is purified using proprietary chromatographic techniques .

Biological Functions

LRG1 plays a significant role in various physiological and pathological processes. It is constitutively expressed by hepatocytes and neutrophils, and its expression is induced in response to various inflammatory stimuli . LRG1 has been implicated in multiple human conditions, including cancer, diabetes, cardiovascular disease, neurological disease, and inflammatory disorders .

One of the key functions of LRG1 is its involvement in the TGFβ signaling pathway, where it acts as an upstream modifier . This pathway is crucial for regulating cell growth, differentiation, and immune responses. LRG1’s modulation of TGFβ signaling can lead to various effects on different cell types, including epithelial, immune, mesenchymal, and cancer cells .

Role in Disease Pathogenesis

LRG1 has been identified as a pathogenic factor in several diseases. It disrupts the cellular interactions required for the formation and maintenance of mature blood vessels, contributing to a highly hypoxic and immunosuppressive microenvironment . This vasculopathic role of LRG1 makes it a potential therapeutic target for diseases characterized by abnormal vascular function.

In cancer, LRG1 is present abundantly in the tumor microenvironment, where it impedes the delivery of therapeutics by contributing to vascular dysfunction . The development of antibody-drug conjugates (ADCs) targeting LRG1 has shown promise in improving the efficacy of cancer treatments .

Therapeutic Potential

Given its role in disease pathogenesis, LRG1 is being explored as a therapeutic target. Inhibition of LRG1 through gene deletion or function-blocking antibodies has been shown to attenuate disease progression in animal studies . This suggests that targeting LRG1 could be a viable strategy for treating various diseases, including cancer and inflammatory disorders.

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.