LCN2 Rat

Neutrophil Gelatinase Associated Lipocalin/Lipocalin-2 Rat Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to LCN2 in Rats

Lipocalin-2 (LCN2), also known as neutrophil gelatinase-associated lipocalin (NGAL), is a 25-kDa secretory protein that plays multifaceted roles in inflammation, iron homeostasis, and cellular stress responses. In rat models, LCN2 is extensively studied due to its dual roles in protective and pathological processes, ranging from bacterial defense to organ injury . This article synthesizes structural, functional, and experimental data on LCN2 in rats, emphasizing its biological significance and translational potential.

Roles in Inflammation and Immune Response

LCN2 is a critical mediator of innate immunity and inflammation in rats:

Proinflammatory Effects

  • Neutrophil recruitment: Drives neutrophil infiltration in lung inflammation, alcoholic liver disease, and psoriasis via chemotactic signaling .

  • Cytokine amplification: Activates NF-κB and MyD88 pathways in epithelial cells, sustaining proinflammatory cytokine production (e.g., IL-6) .

Anti-inflammatory and Resolution Roles

  • Macrophage polarization: Promotes M2-like macrophage activation, aiding inflammation resolution .

  • Apoptosis regulation: Modulates Bim pathway activity to clear apoptotic neutrophils, preventing chronic inflammation .

Expression in Organ Systems

LCN2 exhibits tissue-specific expression and functions:

OrganExpression PatternFunctional Role
LungsSecreted by alveolar macrophages and epithelial cells during irradiation or infectionBacterial iron chelation, neutrophil recruitment .
LiverMajor acute-phase protein (200-fold serum increase post-turpentine injection)Mediates IL-6-dependent inflammatory response .
TestesLocalized in germ cells (disputed in Sertoli/Leydig cells)Potential roles in gonad development and fertility .
BrainUpregulated in neurons post-stroke"Help-me" signal for glial activation and repair .

Disease Associations and Biomarker Potential

LCN2 is implicated in diverse pathologies, serving as a biomarker or therapeutic target:

Experimental and Clinical Correlations

  • Kidney injury: Elevated serum LCN2 predicts acute kidney failure in rat models .

  • Cardiac hypertrophy: Overexpression in cardiomyocytes reduces proliferation and induces hypertrophy .

  • Neurological damage: Post-ischemic neuronal LCN2 activates microglia phagocytosis and astrocyte GFAP expression .

Diagnostic Utility

  • Serum LCN2 levels correlate with disease severity in sterile abscesses, stroke, and metabolic disorders .

Experimental Models and Research Findings

Key rat models elucidating LCN2 dynamics:

ModelKey FindingsReferences
Lung irradiationLCN2 induction in lung tissue parallels IL-6 and TNF-α upregulation .
Turpentine-induced APRLiver-specific LCN2 mRNA increases 8,738-fold, surpassing α₂-macroglobulin .
Focal cerebral ischemiaNeuronal LCN2 increases 2-fold by day 3, activating microglia phagocytosis .
Intrauterine growth restrictionCardiac LCN2 overexpression reduces mitosis and induces hypertrophy .

Receptor Interactions and Signaling Pathways

LCN2 engages multiple receptors to exert its effects:

ReceptorFunctionBinding Affinity (Kd)Pathway
24p3R (NGALR)Iron transport, apoptosis regulation92 pM (holo-LCN2) STAT3, Bcl-2 modulation .
LRP2Cellular uptake, signaling60 nM MAPK/ERK activation .
SLC22A17Germ cell developmentNot quantifiedReproductive tissue regulation .

Product Specs

Introduction
Lipocalin-2, also known as Neutrophil Gelatinase Associated Lipocalin (NGAL), is part of the lipocalin family. This family comprises 25 proteins, including a1-microglobulin and b-lactoglobulin, known for their ability to bind small lipophilic molecules within their hydrophobic core. This characteristic enables them to function as transporters for substances like retinal, biliverdins, and prostaglandins. Studies suggest that NGAL plays a role in regulating inflammatory responses and is detected in the plasma of stroke patients.
Description
Recombinant Rat LCN2, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 201 amino acids (21-198 a.a). It has a molecular weight of 22.9 kDa. This LCN2 protein is fused to a 23 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The LCN2 protein solution has a concentration of 0.25 mg/ml and is prepared in Phosphate Buffered Saline (pH 7.4) with 20% glycerol.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it's recommended to freeze the solution at -20°C. To further enhance long-term stability, adding a carrier protein (0.1% HSA or BSA) is advisable. It's crucial to avoid repeated freeze-thaw cycles.
Purity
The purity of the LCN2 protein is greater than 95.0%, as determined by SDS-PAGE analysis.
Synonyms
Neutrophil gelatinase-associated lipocalin, NGAL, Alpha-2-microglobulin-related protein, Alpha-2U globulin-related protein, Lipocalin-2, Siderocalin LCN2, p25.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSQDSTQNL IPAPPLISVP LQPGFWTERF QGRWFVVGLA ANAVQKERQS RFTMYSTIYE LQEDNSYNVT SILVRGQGCR YWIRTFVPSS RPGQFTLGNI HSYPQIQSYD VQVADTDYDQ FAMVFFQKTS ENKQYFKVTL YGRTKGLSDE LKERFVSFAK SLGLKDNNIV FSVPTDQCID N.

Q&A

What is LCN2 and what are its key functions in rats?

LCN2 (Lipocalin-2), also known as NGAL (Neutrophil gelatinase-associated lipocalin), is a 25 kDa glycosylated single chain monomer belonging to the lipocalin family of proteins. It functions primarily in binding and transporting small lipophilic molecules . In rats, LCN2 is released by activated neutrophils and can form dimers, small amounts of higher oligomers, and complexes with matrix metalloproteinase 9 (MMP-9, gelatinase B) . LCN2 plays significant roles in the innate immune system by binding siderophores, and its expression dramatically increases during inflammatory responses, cancer development, and in response to ischemic or nephrotoxic kidney injury . In the brain, LCN2 serves as a "help-me" signal released by injured neurons to activate microglia and astrocytes, potentially promoting recovery after injury .

How do LCN2 levels differ across rat tissues under normal and pathological conditions?

Under normal conditions, basal LCN2 levels in rat brain tissue are approximately 1.57±0.2 ng/mg protein . During pathological states such as cerebral ischemia, LCN2 levels in the affected hemisphere can increase significantly, reaching approximately 5-10 ng/mg protein (roughly 0.5-1 μg/mL when considering that rat brain total protein concentration is about 100 mg/mL) . Following focal cerebral ischemia, LCN2 begins to increase in the ischemic hemisphere on day 1 and becomes significantly elevated by approximately 2-fold compared to the contralateral hemisphere by day 3, before gradually decreasing back to baseline levels by day 7 .

In kidney tissue, LCN2 expression is dramatically increased following ischemic or nephrotoxic injury . LCN2 expression can also be upregulated in various epithelial tissues during inflammation or cancer development .

What are the recommended methods for measuring LCN2 in rat samples?

Enzyme-linked immunosorbent assay (ELISA) is the most commonly used and reliable method for quantifying LCN2 in rat samples. Commercially available rat Lipocalin-2/NGAL ELISA kits typically employ a sandwich ELISA approach:

  • A polyclonal rat Lipocalin-2/NGAL antibody is pre-coated onto microplate wells

  • Rat test samples and standards of known Lipocalin-2/NGAL concentration are added to wells

  • A biotinylated rat Lipocalin-2/NGAL polyclonal detection antibody is added to form a sandwich

  • After washing, an enzyme is added that reacts with the antibody-target complex

  • The intensity of the resulting signal is directly proportional to the concentration of LCN2

These ELISA kits are validated for measuring LCN2 in various rat sample types including serum, plasma (particularly heparin plasma), and cell culture media .

What are the critical controls needed when studying LCN2 in rat models?

When designing experiments to study LCN2 in rat models, researchers should incorporate the following controls:

  • Sham-operated controls: Essential for studies involving surgical interventions, such as cerebral ischemia models. For example, in ischemic brain studies, sham-operated rats showed LCN2 levels of 1.57±0.2 ng/mg protein .

  • Contralateral tissue controls: When studying focal injuries or regional effects, the unaffected contralateral side serves as an important internal control, as demonstrated in focal cerebral ischemia studies .

  • Temporal controls: Given that LCN2 expression changes dynamically over time (increasing by day 1 after injury, peaking at day 3, and returning to baseline by day 7 in ischemia models), time-course measurements are crucial .

  • Antibody validation controls: Given the documented issues with antibody specificity for LCN2 receptors, experiments should include appropriate negative controls without primary antibodies and validation using multiple antibodies. For instance, neuronal staining for LCN2 was confirmed with three different antibodies in ischemia research .

How should researchers approach immunohistochemical detection of LCN2 in rat tissues?

When performing immunohistochemical detection of LCN2 in rat tissues, researchers should follow these methodological recommendations:

  • Tissue preparation: Transcardially perfuse rats with ice-cold PBS, followed by preparation of fresh-frozen sections (typically 20-μm coronal sections for brain tissue) .

  • Multiple antibody validation: Use at least 2-3 different validated antibodies against LCN2 to confirm specificity of staining patterns. This approach has been successfully employed in studies examining LCN2 in rat brain tissues .

  • Co-localization studies: Perform double or triple labeling with cell-specific markers (e.g., NeuN for neurons, GFAP for astrocytes, Iba1 for microglia) to determine the cellular localization of LCN2 .

  • Negative controls: Always include negative controls without primary antibodies to confirm absence of non-specific staining .

  • Quantification methods: Systematically assess LCN2 immunoreactivity in multiple fields (e.g., peri-infarct fields at 200× magnification) and in corresponding contralateral areas for proper comparison .

What sample preparation methods are optimal for detecting LCN2 in rat blood and tissues?

For optimal detection of LCN2 in rat samples, the following preparation methods are recommended:

Blood samples:

  • Collect blood in appropriate anticoagulant tubes (heparin plasma has been validated for ELISA-based detection)

  • Process samples promptly to prevent degradation

  • Centrifuge at 2,000-3,000 × g for 15-20 minutes to separate plasma or allow blood to clot for serum collection

  • Aliquot and store samples at -80°C to avoid repeated freeze-thaw cycles

Tissue samples:

  • For brain tissue: Transcardial perfusion with ice-cold PBS followed by immediate freezing or fixation

  • For other tissues: Rapid dissection and flash freezing in liquid nitrogen

  • Homogenize tissues in appropriate buffer (typically PBS with protease inhibitors)

  • Clarify homogenates by centrifugation (typically 10,000-15,000 × g for 10-15 minutes at 4°C)

  • Normalize protein concentration across samples before analysis

Cell culture samples:

  • Collect conditioned media from cultured cells (primary cultures or cell lines)

  • Centrifuge to remove cellular debris (typically 300-500 × g for 5-10 minutes)

  • Analyze immediately or store at -80°C with protease inhibitors

How should researchers address inconsistencies in LCN2 receptor studies in rats?

Researchers face significant challenges when studying LCN2 receptors in rats due to documented inconsistencies, misinterpretations, and false assumptions in the literature. To address these challenges, consider the following approaches:

  • Critical antibody validation: Many antibodies directed against LCN2 receptors produce faulty data. Before proceeding with experiments, validate antibodies thoroughly using:

    • Positive controls with transiently expressed proteins

    • Western blot analysis of tissues known to express the receptor

    • Comparison of results using multiple antibodies against different epitopes

  • Expression system verification: When studying LCN2 receptors such as LRP2, NGALR, or MC4R, verify expression using complementary techniques:

    • mRNA detection (RT-PCR, RNA-seq, or in situ hybridization)

    • Protein detection (validated antibodies, mass spectrometry)

    • Functional assays to confirm receptor activity

  • Address isoform complexity: Consider that apparent inconsistencies might result from expression of different receptor isoforms. For example, some tissues might express N-truncated LRP2 isoforms not recognized by certain antibodies .

  • Avoid over-reliance on commercial claims: Be skeptical of commercial antibodies claimed to work across multiple applications and species without rigorous validation evidence .

How can researchers reconcile contradictory findings regarding LCN2's role in metabolism in rat studies?

The literature contains contradictory findings regarding LCN2's role in metabolism, particularly in relation to insulin resistance and obesity. To address these contradictions in rat studies, researchers should:

  • Carefully control experimental conditions: Differences in diet composition, duration of intervention, age of animals, and genetic background can all influence metabolic outcomes.

  • Consider receptor-specific effects: Different receptors for LCN2 may mediate distinct and sometimes opposing metabolic effects. For example:

    • LRP2 acts as an endocytic receptor for insulin reabsorption and shows increased expression in early type 2 diabetes

    • MC4R deficiency induces insulin resistance and obesity without hyperglycemia

    • NGALR's role in metabolism remains incompletely characterized despite its high affinity for LCN2

  • Examine timing and context: LCN2 may have different effects depending on:

    • Acute versus chronic elevation

    • Physiological versus pharmacological concentrations

    • Presence of other inflammatory mediators or stressors

  • Use complementary in vivo and in vitro approaches: Combine whole-animal metabolic phenotyping with cell-specific studies to dissect mechanisms underlying contradictory findings.

What methods should be used to study LCN2's effects on glial cells in rat models?

To properly investigate LCN2's effects on glial cells in rat models, researchers should employ the following methodological approaches:

  • Primary cell culture systems: Isolate and culture primary rat microglia and astrocytes following standard methods to study direct effects of LCN2 .

  • Physiologically relevant concentrations: Based on in vivo measurements from ischemic brain tissue (approximately 0.5-1 μg/mL), use similar concentrations for in vitro experiments .

  • Comprehensive phenotypic analysis for microglia:

    • Morphological assessment (e.g., measuring the length of the longest axis of cells)

    • Gene expression analysis of activation markers (inducible NO synthase, CD86, CD206, arginase 1)

    • Cytokine production measurement (IL-10, IL-1β)

    • Functional assays for phagocytosis and migration

  • Astrocyte-specific assessments:

    • Protein expression analysis for GFAP

    • Growth factor expression (TSP-1, BDNF, VEGF, IGF-1)

    • Morphological analysis

    • Cell viability assessment

  • Conditioned media experiments: Collect conditioned media from LCN2-treated glial cells to assess potential paracrine effects on other cell types .

How reliable are rat models for translating LCN2 findings to human conditions?

Researchers should exercise caution when translating LCN2 findings from rat models to human conditions due to several important considerations:

  • Sequence homology limitations: The percent identity between human and rat LCN2 is only approximately 62%, which is relatively low compared to their receptors that share up to 94% sequence homology. This significant difference raises questions about functional conservation .

  • Demonstrated cross-species differences: Despite the common assumption that orthologs share biological functions, there are reports of functional divergence between mouse and human LCN2 orthologs, which likely extends to rat models as well .

  • Validation in human tissues: When possible, key findings from rat models should be validated in human tissue samples. For example, LCN2 expression in neurons following ischemic stroke was confirmed in postmortem human brain sections, supporting the translational relevance of this particular finding .

  • Systematic translational assessment: Consider that systematic studies investigating animal-to-human translational success rates have shown that translational success is often unpredictable, particularly for inflammatory diseases .

What are the methodological approaches to study LCN2 in rat models of cerebral ischemia?

To effectively study LCN2 in rat models of cerebral ischemia, researchers should employ these methodological approaches:

  • Model selection and standardization:

    • Transient focal cerebral ischemia model (e.g., 90 minutes of occlusion followed by reperfusion)

    • Careful monitoring of physiological parameters during surgery

    • Inclusion of sham-operated controls

  • Temporal assessment:

    • Examine multiple time points (days 1, 3, and 7 post-ischemia) to capture the dynamic changes in LCN2 expression

  • Quantitative measurement:

    • Use ELISA to quantify LCN2 protein levels in brain tissue homogenates

    • Compare ipsilateral (ischemic) hemisphere with contralateral control tissue

  • Cellular localization:

    • Perform multilabel immunostaining with cell-specific markers (NeuN, GFAP, Iba1)

    • Focus on peri-infarct cortical regions where LCN2 expression is most pronounced

  • Functional significance assessment:

    • Conduct in vitro experiments with physiologically relevant concentrations (0.5-1 μg/mL) of LCN2 on primary neural cells

    • Evaluate morphological and molecular changes in glial cells exposed to LCN2

What approaches can address antibody reliability issues in LCN2 receptor research?

Given the documented concerns about antibody reliability in LCN2 receptor research, researchers should implement the following approaches:

  • Comprehensive antibody validation strategy:

    • Test antibodies on cells transiently overexpressing the receptor of interest

    • Use multiple antibodies targeting different epitopes of the same receptor

    • Include genetic models (knockout controls) whenever possible

  • Independent verification methods:

    • Confirm protein expression using mass spectrometry-based proteomics

    • Verify mRNA expression using RT-PCR, RNA-Seq, or in situ hybridization

    • Employ reporter gene constructs to monitor receptor expression

  • Functional validation:

    • Complement immunological detection with functional assays that verify receptor activity

    • Use receptor antagonists or agonists to confirm specificity of observed effects

    • Implement receptor knockdown or knockout approaches to confirm antibody specificity

  • Transparent reporting:

    • Document all antibody validation steps in publications

    • Report negative results regarding antibody performance to prevent propagation of unreliable reagents

    • Include detailed methods for antibody validation, including catalog numbers, dilutions, and specific protocols used

How can the sensitivity and specificity of LCN2 detection be optimized in rat experimental samples?

To optimize the sensitivity and specificity of LCN2 detection in rat experimental samples:

  • ELISA optimization:

    • Use validated, rat-specific sandwich ELISA kits with documented sensitivity, specificity, precision, and lot-to-lot consistency

    • Follow manufacturer's protocols precisely, paying particular attention to incubation times and temperatures

    • Generate standard curves in the same matrix as experimental samples (e.g., serum, plasma, cell culture media)

    • Run samples in duplicate or triplicate to ensure reproducibility

  • Sample handling improvements:

    • Process samples immediately after collection to prevent degradation

    • Add protease inhibitors to tissue homogenates and cell lysates

    • Avoid repeated freeze-thaw cycles by storing samples in single-use aliquots

    • Ensure consistent protein determination methods across all samples

  • Immunohistochemistry enhancement:

    • Optimize fixation protocols for specific tissues

    • Use antigen retrieval techniques appropriate for the epitope being detected

    • Employ tyramide signal amplification for low abundance targets

    • Use confocal microscopy for precise cellular localization

What are the best approaches for studying LCN2 receptor dynamics in rat models?

To effectively study LCN2 receptor dynamics in rat models:

  • Receptor expression analysis:

    • Quantify receptor mRNA levels using qRT-PCR with validated primer sets

    • Assess protein expression using thoroughly validated antibodies against specific LCN2 receptors (LRP2, NGALR, MC4R)

    • Map tissue-specific expression patterns using in situ hybridization combined with immunohistochemistry

  • Receptor internalization and trafficking studies:

    • Use fluorescently labeled LCN2 to track receptor-mediated internalization

    • Employ live-cell imaging to monitor receptor trafficking in real-time

    • Utilize subcellular fractionation followed by immunoblotting to assess receptor localization

  • Signaling pathway analysis:

    • Investigate downstream signaling events following LCN2 binding to specific receptors

    • Study phosphorylation of key signaling molecules at multiple time points

    • Use pathway-specific inhibitors to delineate signaling cascades activated by LCN2

  • Genetic approaches:

    • Implement receptor knockdown using siRNA or shRNA in cell culture systems

    • Consider CRISPR/Cas9-mediated receptor knockout in rat cell lines

    • Use tissue-specific conditional knockout models to study receptor function in specific cell populations

How can researchers differentiate between direct and indirect effects of LCN2 in rat neuroinflammation studies?

Differentiating between direct and indirect effects of LCN2 in rat neuroinflammation studies requires careful experimental design:

  • Cell-specific isolation and treatment:

    • Isolate primary neurons, microglia, and astrocytes from rat brains

    • Treat each cell type independently with purified LCN2

    • Document cell-specific responses using appropriate marker analyses

  • Conditioned media experiments:

    • Collect conditioned media from LCN2-treated microglia or astrocytes

    • Apply this conditioned media to other cell types (e.g., neurons)

    • Compare effects of direct LCN2 application versus LCN2-conditioned media

  • Receptor antagonism or genetic manipulation:

    • Use selective antagonists for specific LCN2 receptors

    • Implement receptor knockdown approaches in specific cell types

    • Compare effects in wild-type versus cells with receptor modification

  • In vivo approaches:

    • Use cell-specific genetic deletion of LCN2 receptors

    • Implement in vivo microdialysis to deliver LCN2 or receptor antagonists locally

    • Combine with in vivo imaging techniques to monitor cellular responses in real-time

Product Science Overview

Introduction

Neutrophil Gelatinase Associated Lipocalin (NGAL), also known as Lipocalin-2 (LCN2), is a protein that plays a crucial role in the body’s innate immune response. It was first identified in a simian virus 40-infected murine kidney cell culture and later found to be associated with matrix metalloproteinase (MMP)-9, a gelatinase secreted by neutrophils for extracellular matrix remodeling . NGAL is involved in iron homeostasis, infection, and inflammation, making it a significant protein in various physiological and pathological processes .

Functions

NGAL is primarily known for its ability to bind and sequester iron, which is essential for bacterial growth. By binding to bacterial siderophores, NGAL limits the availability of iron to bacteria, thereby inhibiting their growth . This function is crucial in the body’s defense against bacterial infections. Additionally, NGAL binds to the mammalian siderophore 2,5-dihydroxybenzoic acid (2,5-DHBA), ensuring that excess free iron does not accumulate in the cytoplasm, which could lead to high levels of reactive oxygen species .

Clinical Significance

NGAL is used as a biomarker for kidney injury due to its protease resistance and low molecular weight, which allows it to be excreted and detected in urine . Injured epithelial cells in the kidney secrete a monomeric form of NGAL, while activated neutrophils secrete a dimeric form. This distinction can potentially improve acute kidney injury (AKI) diagnostics by differentiating between NGAL of inflammatory origin and that of renal origin . Elevated NGAL levels are observed in both blood and urine within two hours of kidney injury, making it a more precise and sensitive marker for diagnosing AKI compared to serum creatinine levels .

Role in Sepsis

NGAL has also been recognized for its roles in gut-origin sepsis. It mitigates gut barrier injury by maintaining microbiota homeostasis, exerting antioxidant effects, deactivating macrophages, and inducing immune cell apoptosis to terminate systemic hyper-inflammation . This makes NGAL a promising therapeutic target in the management of gut-origin sepsis.

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.