Lcn2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Lcn2 Antibody

Lcn2 antibodies are immunoglobulins specifically designed to recognize and bind to Lipocalin-2 (Lcn2), also commonly known as Neutrophil Gelatinase-Associated Lipocalin (NGAL). These antibodies are extensively used in laboratory and clinical settings for various applications including protein detection, quantification, and neutralization. As research tools, they enable scientists to understand the diverse biological functions of Lcn2 and its role in various disease states .

Lcn2 antibodies are commercially available in various formats including monoclonal and polyclonal variants with different species reactivity profiles. These antibodies are validated for multiple applications such as Western blotting, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), and flow cytometry, making them versatile tools for both basic research and clinical diagnostics .

Biological Functions

Lcn2 serves multiple physiological functions:

  1. Iron trafficking and homeostasis: Binds iron through association with 2,5-dihydroxybenzoic acid (2,5-DHBA), a siderophore that shares structural similarities with bacterial enterobactin

  2. Innate immunity: Acts as an antibacterial agent by sequestering iron that limits bacterial growth

  3. Cell differentiation and apoptosis: Involved in regulating cellular apoptosis through iron-dependent mechanisms

  4. Neutrophil function: Essential for neutrophil recruitment, migration, adhesion, and function

  5. Tissue development: Plays a role in renal development

Expression Profile

Lcn2 demonstrates a specific expression pattern across tissues and cells:

Tissue/Cell TypeExpression LevelReference
NeutrophilsHigh (protein level)
Bone marrowVery high
UterusHigh
ProstateHigh
Salivary glandHigh
StomachHigh
AppendixHigh
ColonHigh
TracheaHigh
LungHigh
Small intestineNot detected
Peripheral blood leukocytesNot detected

Lcn2 expression is significantly upregulated during inflammation, infection, and injury conditions .

Classification Based on Source/Host

Lcn2 antibodies are derived from various host animals, each offering distinct advantages:

HostTypeExamplesApplicationsReference
RabbitRecombinant MonoclonalEPR5084 (ab125075)WB, IHC-P, ICC/IF
RabbitPolyclonalDF6816WB, IHC
RatMonoclonal220310, 220322WB, IHC, ELISA

Classification Based on Reactivity

Lcn2 antibodies vary in their species reactivity profiles:

ReactivityExample ProductsApplicationsReference
Human-specificab125075, MAB1757WB, IHC, ICC/IF
Human/Mouse/RatAF1757WB, IHC
Human, Mouse, Rat, OthersDF6816, bs-1373RWB, IHC, ELISA, FCM

Classification Based on Applications

Lcn2 antibodies are optimized for specific laboratory techniques:

ApplicationRecommended DilutionExample ProductsReference
Western Blot (WB)1:1,000-1:5,000ab125075, MAB1757, A00452-1
Immunohistochemistry (IHC)1:50-1:200MAB1757, A00452-1
Immunocytochemistry (ICC)1:100-1:500ab125075, A00452-1
ELISAVaries by productMAB17571
Flow Cytometry (FCM)1:50-1:100bs-1373R

Diagnostic Applications

Lcn2 antibodies have demonstrated significant utility in diagnostic applications across multiple fields:

  1. Cancer Biomarker Detection: Detecting elevated Lcn2 levels in various cancers including:

    • Cervical cancer - Higher levels correlate with advanced clinical stages

    • Inflammatory breast cancer - Aberrantly abundant compared to non-inflammatory breast cancer

    • Pancreatic adenocarcinoma - Detected in Capan-1 cell line

  2. Inflammatory Disease Assessment: Monitoring Lcn2 levels in:

    • Systemic lupus erythematosus (SLE)

    • Arthritis

    • Inflammatory bowel disease - Expression in colon tissue

  3. Renal Function Evaluation: Lcn2/NGAL serves as a biomarker for acute kidney injury

Research Applications

In laboratory settings, Lcn2 antibodies enable critical research applications:

  1. Protein Detection and Quantification:

    • Western blot analysis of Lcn2 expression in cell lysates and tissue samples

    • ELISA-based quantification of Lcn2 levels in biological fluids

    • Immunohistochemical localization of Lcn2 in tissue sections

  2. Functional Studies:

    • Investigation of Lcn2's role in iron homeostasis

    • Analysis of Lcn2's contribution to inflammatory responses

    • Elucidation of Lcn2's involvement in neutrophil function

  3. Knockout Validation Studies:

    • Verification of Lcn2 knockout models - as demonstrated by the loss of signal in LCN2 knockout cell lines

    • Comparative studies between wild-type and Lcn2 knockout systems

Therapeutic Applications

Emerging research highlights the therapeutic potential of Lcn2 antibodies:

  1. Cancer Treatment:

    • Anti-Lcn2 antibody treatment decreased proliferation of cervical cancer cells (HeLa) in a dose-dependent manner

    • Reduced invasive ability of cancer cells in a dose-dependent manner

    • Potential target against inflammatory breast cancer

  2. Inflammatory Disease Management:

    • Neutralization of Lcn2 using monoclonal antibodies significantly reduced inflammation in wild-type mice

    • Potential therapeutic approach for immune-complex mediated inflammatory diseases

  3. Ocular Disease Treatment:

    • Neutralizing Lcn2 with specific antibodies could alleviate autophagy abnormalities and redox imbalance in retinal pigment epithelium (RPE) cells

    • Potential strategy for age-related macular degeneration (AMD) treatment

Validation Studies

Lcn2 antibodies have undergone rigorous validation across multiple platforms:

  1. Western Blot Validation:

    • Ab125075 successfully detected Lcn2 in wild-type SW480 cells with loss of signal in LCN2 knockout cell line

    • MAB1757 detected specific bands for Lcn2 at approximately 22 kDa in Capan-1 human pancreatic adenocarcinoma cell line and human pancreas tissue

    • AF1757 detected Lcn2 in mouse and rat bone marrow at approximately 22 kDa

  2. Immunohistochemistry Validation:

    • Ab125075 successfully labeled Lcn2 in paraffin-embedded human kidney tissue sections

    • MAB1757 detected Lcn2 in human pancreas, specifically localizing to plasma membrane of ductal cells

  3. ELISA Standard Curve Development:

    • MAB17571 was successfully used to create a standard curve for Lcn2 quantification when paired with MAB17573 as detection antibody

Therapeutic Neutralization Studies

Several studies have investigated the therapeutic potential of Lcn2 antibodies:

  1. Inflammatory Disease Models:

    • In acute model of IC-mediated skin inflammation, neutralization of Lcn2 using monoclonal antibody significantly reduced inflammation in wild-type mice

    • Lcn2 knockout mice exhibited reduced inflammation with histopathological analysis revealing strikingly reduced immune cell infiltration

  2. Cancer Studies:

    • Anti-Lcn2 antibody treatment of HeLa cells reduced:

      • Cell proliferation (starting as early as 12 hours after treatment)

      • Invasive ability in a dose-dependent manner

      • Expression of active metalloproteases (likely MMP9)

    • Treatment increased expression of Claudin mRNA, suggesting restriction in migratory abilities

  3. Ocular Disease Research:

    • Antibody clones 2, 3, and 11 showed binding with both Lcn2 monomer and homodimer

    • Cryba1 KO RPE explant cultures treated with these clones showed reduced levels of Lcn2 compared to untreated explants

Future Directions in Lcn2 Antibody Research and Applications

The expanding knowledge of Lcn2's roles in various diseases opens new avenues for antibody applications:

  1. Novel Therapeutic Strategies:

    • Development of humanized anti-Lcn2 antibodies for clinical applications

    • Combination therapies with existing treatments

    • Targeted delivery systems for improved efficacy

  2. Advanced Diagnostic Applications:

    • Development of point-of-care testing for Lcn2 levels

    • Integration with other biomarkers for improved disease detection

    • Personalized medicine approaches based on Lcn2 expression profiles

  3. Expanded Research Tools:

    • Creation of bispecific antibodies targeting Lcn2 and its binding partners

    • Development of antibodies specific to different Lcn2 forms (monomer vs. dimer)

    • Engineering of intrabodies for intracellular targeting of Lcn2

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
Lcn2 antibody; Neutrophil gelatinase-associated lipocalin antibody; NGAL antibody; Alpha-2-microglobulin-related protein antibody; Alpha-2U globulin-related protein antibody; Lipocalin 24p3 antibody; Lipocalin-2 antibody; Siderocalin LCN2 antibody; p25 antibody
Target Names
Uniprot No.

Target Background

Function

Lipocalin 2 (LCN2), also known as Neutrophil Gelatinase-Associated Lipocalin (NGAL), is an iron-trafficking protein implicated in diverse biological processes, including apoptosis, innate immunity, and renal development. It binds iron via association with 2,3-dihydroxybenzoic acid (2,3-DHBA), a siderophore structurally similar to bacterial enterobactin. LCN2's role in iron homeostasis is context-dependent; it can either deliver or remove iron from cells. The iron-bound form (holo-24p3) is internalized after binding to the SLC22A17 (24p3R) receptor, resulting in iron release and increased intracellular iron concentration. Conversely, the iron-free form (apo-24p3) binds to SLC22A17 (24p3R), subsequently associating with an intracellular siderophore, chelating iron, and transferring it extracellularly, thus reducing intracellular iron levels. LCN2's involvement in apoptosis is demonstrated by its modulation of intracellular iron concentration following interleukin-3 (IL3) deprivation. The iron-loaded form increases intracellular iron without inducing apoptosis, whereas the iron-free form decreases intracellular iron, upregulating the proapoptotic protein BCL2L11/BIM and triggering apoptosis. LCN2 contributes to innate immunity by sequestering iron from microbial siderophores, like enterobactin, thereby limiting bacterial proliferation. It also exhibits binding affinity for siderophores from Mycobacterium tuberculosis.

Gene References Into Functions

Further Research Supporting LCN2 Function:

  • NGAL's potential role in mitigating tubular epithelial cell damage through autophagy modulation. PMID: 29367586
  • Remote limb preconditioning's effect on LCN2 protein expression following cerebral ischemia. PMID: 29122651
  • The association between high LCN2 expression, stress-induced mitochondrial dysfunction, and heart failure. PMID: 29378951
  • Investigation into NGAL levels and autophagy during renal ischemia/reperfusion injury. PMID: 29704511
  • The correlation between NGAL, microglial activity, and postoperative behavioral changes in rats. PMID: 27865916
  • Marked increases in LCN-2 protein levels in the ipsilateral cortex and hippocampus after traumatic brain injury, primarily within astrocytes. PMID: 26463963
  • Nitric oxide's influence on LCN-2 expression and its potential role in beta-cell apoptosis. PMID: 27233602
  • NGAL upregulation in nerve degeneration and its potential role in nerve repair. PMID: 27087673
  • LCN2 as a potential biomarker for lung hypoplasia in congenital diaphragmatic hernia (CDH). PMID: 27592368
  • Synergistic effects of hypertension and hyperglycemia on renal tubular alterations and increased urinary NGAL excretion. PMID: 25148248
  • The role of the Pla2g4c gene in mammary carcinogenesis and its relationship to NF-kappaB/Lcn2-induced apoptosis. PMID: 26013918
  • Epidural anesthesia's impact on lipocalin-2 and E. coli DNA expression in rats. PMID: 25782996
  • Dose- and time-dependent increase in KIM-1 and NGAL expression following repeated gentamicin administration in rats. PMID: 25087119
  • Dietary fructose's contribution to lipocalin-2 overexpression in rat fatty liver. PMID: 24587658
  • LCN2's regulatory role in small-for-size fatty liver graft injury and its potential as a therapeutic target. PMID: 24374540
  • NGF-induced LCN2 expression in intervertebral disk cells and its inhibition by Ro 08-2750. PMID: 25062286
  • Lipocalin-2's role as a distress signal released by injured neurons, activating microglia and astrocytes. PMID: 24916903
  • The r24p3-R/hNGAL-R receptor's potential role in protein/peptide internalization and transcytosis in the lower intestine. PMID: 23940770
  • Comparison of urinary KIM-1, NGAL, and NAG with serum creatinine and renal histological scores in cisplatin-induced acute kidney injury. PMID: 23360846
  • LCN2 gene expression during liver regeneration after partial hepatectomy. PMID: 23481292
  • The correlation between NGAL expression, renal interstitial fibrosis, and markers of inflammation (MMP-9, TNF-alpha, TGF-beta1). PMID: 23158800
  • LCN2 induction in activated astrocytes in response to neurodegeneration, independent of TDP-43 or FUS expression. PMID: 23431168
  • The association between high NGAL levels and proteinuria. PMID: 21551085
  • The relationship between acute myocardial infarct in chronic kidney disease, accelerated renal fibrosis, and elevated urinary NGAL. PMID: 22889806
  • Renal changes in rats exposed to methyl tertiary-butyl ether, suggestive of alpha(2u)-globulin nephropathy. PMID: 22833177
  • The role of IL-10 macrophages and lipocalin-2 in attenuating acute kidney injury. PMID: 22278021
  • LIPOCALIN-2 as a major acute-phase protein in a rat and mouse model of sterile abscess. PMID: 22249220
  • LCN2-induced cardiomyocyte apoptosis via a mechanism involving elevated intracellular iron and Bax translocation. PMID: 22117066
  • SIP24's modulation of neutrophil populations through apoptosis induction. PMID: 16546827
  • NGAL's interaction with MMP-9 and its potential role in vascular repair. PMID: 17148685
  • The correlation between LCN2 expression, liver damage, inflammatory responses, and liver fibrosis. PMID: 21457438
  • LCN2/NGAL expression in cardiomyocytes, vascular wall cells, and fibroblasts during myocarditis. PMID: 20057160
  • LCN2 induction in the lung and liver during the acute phase response to acute endotoxemia. PMID: 17490638
Database Links
Protein Families
Calycin superfamily, Lipocalin family
Subcellular Location
Secreted. Cytoplasmic granule lumen. Cytoplasmic vesicle lumen.
Tissue Specificity
Detected in the ureteric bud in embryonic kidney (at protein level).

Q&A

What cell types express Lcn2 and where should researchers focus their immunostaining studies?

Lcn2 expression has been identified in multiple cell types involved in inflammatory and immune responses. Research indicates that neutrophils, astrocytes, and vascular endothelial cells strongly express Lcn2, particularly under pathological conditions. In brain ischemia models (tMCAo), Lcn2 has been detected in astrocytes whose end-feet encircle blood vessels, neutrophils both within blood vessels and in ischemic brain parenchyma, and on the inner surface of vascular endothelial cells . When designing immunostaining experiments, researchers should include markers for these specific cell types (e.g., GFAP for astrocytes, Ly-6B.2 clone 7/4 for neutrophils, and tomato lectin for blood vessels) to properly identify Lcn2-expressing cells in your tissue of interest.

How does Lcn2 expression change during inflammatory conditions?

Lcn2 levels are significantly elevated across multiple autoimmune disease models, including systemic lupus erythematosus (SLE), collagen-induced arthritis, and serum-induced arthritis . Experimental evidence from mouse models shows that Lcn2 mRNA levels become significantly induced after transient middle cerebral artery occlusion (tMCAo) compared to naive conditions . When designing experiments to study Lcn2 in inflammatory contexts, time-course studies are essential to capture peak expression, which typically occurs within 23 hours after inflammatory insult in acute models . Quantitative analysis methods such as ELISA for protein levels and real-time RT-PCR for mRNA expression provide complementary data to understand the dynamics of Lcn2 upregulation.

What controls should be included when validating Lcn2 antibody specificity?

Proper validation of Lcn2 antibody specificity requires several controls. Experiments using Lcn2 knockout (Lcn2KO) mice tissues provide the gold standard negative control, as demonstrated in multiple studies that show complete absence of signal in these tissues . Immunoprecipitation experiments can further validate specificity by showing that the Lcn2 monoclonal antibody (mAb) can specifically immunoprecipitate both recombinant and endogenous Lcn2 proteins . For Western blot applications, researchers should run parallel samples from wild-type and Lcn2KO mice alongside recombinant Lcn2 protein to confirm band specificity at the expected molecular weight (approximately 37 kDa under reducing conditions) .

How should researchers interpret contradictory findings on Lcn2's role in different inflammatory models?

Lcn2 demonstrates context-dependent roles in inflammatory conditions that may appear contradictory. For instance, in acute skin inflammation models, Lcn2KO mice show approximately 50% reduction in inflammation with notably reduced immune cell infiltration compared to wild-type mice . Conversely, in serum-induced arthritis, Lcn2KO mice develop more severe disease with extensive tissue and bone destruction . These divergent outcomes can be attributed to tissue-specific functions of Lcn2 and differential effects on immune cell populations. When designing experiments to study Lcn2 function, researchers should carefully characterize neutrophil versus macrophage infiltration, as Lcn2KO mice show reduced neutrophil infiltration but increased macrophage migration in arthritis models . Multiple inflammation parameters should be measured, including tissue destruction, cellular infiltration, and inflammatory mediator production to fully understand Lcn2's multifaceted roles.

What are the methodological considerations when using Lcn2 antibodies to study iron sequestration mechanisms?

Lcn2's bacteriostatic properties stem from its ability to sequester bacterial iron siderophores, making iron homeostasis studies crucial to understanding its function . When designing experiments to investigate this mechanism, researchers should include conditions that manipulate iron availability. Studies in Lcn2-deficient mice have confirmed that neutrophils lacking Lcn2 are less effective at inhibiting bacterial growth, especially under iron-limiting conditions . Experimental designs should include bacterial growth assays comparing wild-type and Lcn2KO neutrophils under both iron-replete and iron-depleted conditions. Additionally, researchers should consider examining changes in the microbiome composition, as Lcn2 deficiency has been shown to promote expansion of siderophore-dependent bacterial species, affecting gastrointestinal tract inflammation .

How can researchers effectively use Lcn2 antibodies to investigate its role in infectious disease models?

To investigate Lcn2's role in infectious disease, a multifaceted approach is required. Researchers should utilize Lcn2 antibodies for both neutralization experiments and detection purposes. Neutralization studies have demonstrated that administration of monoclonal antibodies against Lcn2 significantly reduces inflammation in wild-type mice, directly implicating Lcn2 in the inflammatory process . For mechanistic studies, comparing bacterial susceptibility between wild-type and Lcn2KO mice under various iron conditions provides insights into Lcn2's bacteriostatic functions . When designing these experiments, researchers should consider both acute and chronic infection models, as Lcn2's role may differ based on infection duration and pathogen type. Immunohistochemical staining should be performed to track neutrophil infiltration and Lcn2 expression at infection sites, with co-localization studies using markers for immune cells and bacteria.

What are the optimal protocols for detecting Lcn2 expression in tissue sections using immunohistochemistry?

For successful immunohistochemical detection of Lcn2 in tissue sections, several methodological considerations are crucial. Based on published protocols, researchers should perform tissue fixation with 4% paraformaldehyde, followed by cryosectioning for optimal antigen preservation. For multiplex immunofluorescence staining, use Lcn2 antibody (such as AF1857) at a concentration of 5 μg/mL , along with cell-type specific markers to identify Lcn2-expressing cells. For brain tissue, combining Lcn2 antibody (labeled green) with tomato lectin (red, for blood vessels) and GFAP antibody (blue, for astrocytes) enables identification of cell-specific expression patterns . Nuclear counterstaining with DAPI further enhances cell identification. Image acquisition should be performed using confocal microscopy with appropriate filter settings to minimize bleed-through between fluorescence channels. For quantification, analyze at least 5 samples per group, and report the percentage of Lcn2-positive cells by type, as demonstrated in previous studies .

What is the most reliable method for quantifying Lcn2 protein levels in biological samples?

For reliable quantification of Lcn2 protein levels in biological samples, ELISA represents the gold standard method. When analyzing brain tissue, homogenization should be performed in RIPA buffer with protease inhibitors, followed by centrifugation to obtain clear supernatant. For blood samples, serum separation should be performed promptly following collection to prevent ex vivo changes in Lcn2 levels . Research indicates that ELISA can effectively detect changes in Lcn2 concentration in both ipsilateral hemispheres of brain tissue and blood sera following experimental manipulations such as tMCAo or antibody treatment . When reporting results, data should be expressed as concentration (ng/ml) with appropriate statistical analysis comparing experimental groups (n=5-10 per group recommended). For statistical robustness, unpaired t-tests (one-tailed for directional hypotheses, two-tailed for non-directional hypotheses) should be employed when comparing two groups .

How should Western blot protocols be optimized for detecting Lcn2 in different sample types?

Western blot detection of Lcn2 requires optimization based on sample type and experimental context. For mouse lung tissue lysates, loading at 0.2 mg/mL concentration has proven effective . Samples should be prepared under reducing conditions for optimal detection. When probing for Lcn2, researchers should use 5 μg/mL of anti-Lcn2 antibody (such as AF1857) followed by appropriate HRP-conjugated secondary antibody (e.g., 1:50 dilution of anti-goat IgG secondary) . Importantly, Lcn2 typically appears at approximately 37 kDa when using a 12-230 kDa separation system . For immunoprecipitation experiments, incubate increasing concentrations of Lcn2 mAb (0, 0.1, 0.5, and 2.5 μg) bound to magnetic beads with your protein sample, followed by Western blot analysis to confirm specific pull-down of Lcn2 . Always include appropriate positive controls (recombinant Lcn2 protein) and negative controls (samples from Lcn2 knockout animals) to validate specificity.

How can researchers differentiate between free and siderophore-bound forms of Lcn2 in experimental samples?

Differentiating between free and siderophore-bound forms of Lcn2 presents a significant challenge in experimental settings. These forms play distinct roles in Lcn2's bacteriostatic function through iron sequestration . To distinguish between these forms, researchers should employ a combination of analytical approaches. Immunoprecipitation with anti-Lcn2 antibodies followed by mass spectrometry can identify bound siderophores. Additionally, size-exclusion chromatography can separate the different molecular weight complexes prior to Western blot analysis. When interpreting results, researchers should note that Lcn2's bacteriostatic properties are primarily observed under iron-limiting conditions, where it can sequester bacterial siderophores . Comparative analysis between wild-type and Lcn2-deficient models under varying iron conditions provides further insights into the functional relevance of these different forms. Fluorescence-based binding assays using labeled siderophores can also quantify the binding capacity of Lcn2 in experimental samples.

What factors contribute to variability in Lcn2 detection across different experimental models?

Several factors contribute to variability in Lcn2 detection across experimental models. Temporal dynamics of Lcn2 expression show significant changes after inflammatory stimuli, with peak expression typically occurring within 23 hours in acute models . Sample collection timing is therefore critical. The anatomical location of inflammation also influences expression patterns; in brain ischemia models, Lcn2 expression varies between vascular endothelial cells, neutrophils, and astrocytes . Additionally, the inflammatory stimulus type substantially impacts expression levels, with immune-complex mediated inflammation showing distinct patterns from infectious stimuli . Genetic background of experimental animals can also contribute to variability, necessitating proper controls including littermates. Technical factors such as antibody clone selection, detection method sensitivity, and sample processing protocols further contribute to inter-laboratory variation. To minimize variability, researchers should standardize sample collection timing, processing methods, and employ multiple detection techniques (e.g., combining protein detection via ELISA with mRNA quantification via RT-PCR) .

How should researchers interpret differences between mRNA and protein levels of Lcn2 in their experimental systems?

Discrepancies between Lcn2 mRNA and protein levels are commonly observed and require careful interpretation. These differences may reflect several biological processes including post-transcriptional regulation, protein stability differences, or compartmentalization effects. Evidence from tMCAo models shows that while Lcn2 mRNA levels increase significantly after ischemic insult, protein levels may show different patterns in tissue versus serum . When designing experiments, researchers should perform parallel analyses of mRNA (via real-time RT-PCR) and protein (via ELISA or Western blot) from the same experimental groups . For accurate interpretation, consider that antibody-based neutralization of Lcn2 protein may affect observed protein levels without necessarily impacting mRNA expression directly. In studies with Lcn2 mAb treatment, researchers have observed significant decreases in Lcn2 protein concentration in both brain tissue and serum while also noting reduced mRNA expression, suggesting potential feedback mechanisms . Time-course studies tracking both mRNA and protein can help elucidate the relationship between transcription, translation, and protein turnover in your specific experimental system.

How can researchers utilize Lcn2 antibodies to investigate interactions between Lcn2 and its receptors?

Investigating interactions between Lcn2 and its receptors requires sophisticated methodological approaches. Two main receptors have been identified: murine NGALR (discovered through expression cloning in COS-7 cells using a cDNA library from murine FL5.12 cells) and LRP2 (first identified in rats through antibody screening) . To study these interactions, researchers should employ co-immunoprecipitation approaches using anti-Lcn2 antibodies to pull down receptor complexes from cellular lysates, followed by receptor-specific detection methods. Surface plasmon resonance (SPR) or biolayer interferometry using purified components can determine binding kinetics and affinities between Lcn2 and its receptors. For cellular studies, immunofluorescence co-localization experiments using antibodies against both Lcn2 and its receptors can visualize interaction sites within tissues or cells . Proximity ligation assays provide another powerful tool to visualize protein-protein interactions in situ. Functional studies comparing wild-type, receptor knockout, and Lcn2 knockout models help elucidate the physiological relevance of these interactions in various contexts, particularly in iron homeostasis and inflammatory conditions.

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.