LRP1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Production Methodology

Recombinant monoclonal antibodies (mAbs) are synthesized using in vitro expression systems rather than traditional hybridoma techniques. The process involves:

  1. Cloning: Antibody DNA sequences from immunoreactive rabbits are cloned into expression vectors.

  2. Screening: Individual clones are tested for target binding and specificity.

  3. Production: Selected clones are expressed in bacterial or mammalian systems, yielding homogeneous antibodies.

Advantages include:

  • Consistency: Lot-to-lot uniformity.

  • Broad Reactivity: Enhanced immunoreactivity due to diverse rabbit immune repertoires.

  • Ethical Compliance: Animal origin-free formulations .

Specificity and Reactivity

Antibody CloneHostReactivityEpitope TargetSource
FAB6360AMouseHuman, No cross-reactivity with LRP1 clusters 2, 3, 4Ectodomain (Gln4449-Ala4544)
E-AB-81579RabbitHuman, Mouse, RatSynthetic peptide (C-terminal)
EPR3724RabbitHuman, Mouse, Rat, PigFull-length LRP1
SA0290RabbitHuman, Mouse, RatNot specified

Epitope Targeting

Most antibodies target the ectodomain or intracellular domain of LRP1:

  • Ectodomain: Detects surface-expressed LRP1 (e.g., FAB6360A, MAB6360) .

  • Intracellular Domain: Identifies cytoplasmic/nuclear LRP1 (e.g., MAB6360) .

Western Blotting

AntibodyDilutionObserved Band SizeSample TypeReference
E-AB-815791:1000–1:200085 kDaRat Brain, Human Cholangiocarcinoma
EPR37241/5000–1/10,000504 kDa (full-length)HAP1 Cell Lysates
MAB63603 µg/mLIntracellular LRP1HepG2 Cells

Key Findings:

  • Full-Length Detection: EPR3724 identifies the 504 kDa mature LRP1 in wild-type cells, with loss of signal in LRP1 knockout models .

  • Isoform Detection: E-AB-81579 detects processed 85 kDa subunits in rat brain tissue .

Flow Cytometry

  • Surface Expression: FAB6360A detects LRP1 on HEK293 cells, confirming surface localization .

  • Intracellular Staining: EPR3724 labels LRP1 in Jurkat T cells, enabling analysis of cytoplasmic/nuclear pools .

Immunohistochemistry (IHC)

AntibodyDilutionTissueStaining PatternReference
EPR37240.26 µg/mLHuman TestisCytoplasmic
MAB63605 µg/mLHuman LiverHepatocyte cytoplasm

Key Observations:

  • Tissue-Specific Localization: LRP1 is abundant in liver hepatocytes and testicular cells .

Immune Regulation

  • Graft-Versus-Host Disease (GVHD): LRP1 deletion in T cells reduces GVHD severity by lowering proinflammatory cytokines (TNFα, IFNγ) and enhancing Treg populations .

  • Antigen Presentation: LRP1 on APCs enhances T/B cell responses by facilitating pathogen antigen uptake .

Neurodegenerative Diseases

  • Alzheimer’s Disease: LRP1 mediates clearance of β-amyloid plaques. Antibodies like EPR3724 may aid in studying amyloid precursor protein (APP) metabolism .

  • Tauopathies: LRP1 regulates TAU/MAPT endocytosis, impacting neurodegenerative spread .

Cancer and Metabolism

  • Tumor Growth: Inhibiting LRP1 reduces cancer cell proliferation and invasion in gastrointestinal tumors .

  • Lipoprotein Metabolism: LRP1 modulates LDL and chylomicron remnant clearance, influencing atherosclerosis .

Challenges and Future Directions

  • Epitope Variability: Distinct antibodies target different LRP1 domains, necessitating validation for specific assays.

  • Therapeutic Potential: Recombinant antibodies may serve as tools to modulate LRP1 activity in diseases like Alzheimer’s or GVHD.

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
A2MR antibody; Alpha 2 macroglobulin receptor antibody; alpha 2MR antibody; Alpha-2-macroglobulin receptor antibody; APOER antibody; Apolipoprotein E receptor antibody; APR antibody; CD 91 antibody; CD91 antibody; CD91 antigen antibody; IGFBP3R antibody; LDL receptor related protein 1 antibody; Low density lipoprotein receptor related protein 1 antibody; Low density lipoprotein related protein 1 antibody; Low-density lipoprotein receptor-related protein 1 intracellular domain antibody; LRP 1 antibody; LRP 515 antibody; LRP 85 antibody; LRP antibody; LRP ICD antibody; LRP-1 antibody; LRP-515 antibody; LRP-85 antibody; Lrp1 antibody; LRP1 protein antibody; LRP1_HUMAN antibody; LRP1A antibody; LRP515 antibody; LRP85 antibody; LRPICD antibody; MGC88725 antibody; Prolow density lipoprotein receptor related protein 1 antibody; TbetaR V/LRP 1/IGFBP 3 receptor antibody; TbetaRV/LRP1/IGFBP3 receptor antibody; TGFBR 5 antibody; TGFBR5 antibody; Type V tgf beta receptor antibody
Target Names
Uniprot No.

Target Background

Function
LRP1 (Low-density lipoprotein receptor-related protein 1) is an endocytic receptor involved in endocytosis and phagocytosis of apoptotic cells. It is essential for early embryonic development and plays a role in cellular lipid homeostasis. LRP1 is involved in the plasma clearance of chylomicron remnants and activated LRPAP1 (alpha 2-macroglobulin), as well as the local metabolism of complexes between plasminogen activators and their endogenous inhibitors. It serves as an LRPAP1 alpha-2-macroglobulin receptor. LRP1 functions as a TAU/MAPT receptor, controlling the endocytosis of TAU/MAPT and its subsequent spread. It may modulate cellular events such as APP metabolism, kinase-dependent intracellular signaling, neuronal calcium signaling, and neurotransmission. LRP1 also acts as a receptor for Pseudomonas aeruginosa exotoxin A, which can have implications in microbial infections.
Gene References Into Functions
  1. The association of LRP1 rs11613352 and angiopoietin-like 3 rs2131925 with hypertension suggests a potential direct effect at the artery wall. PMID: 29989339
  2. Research indicates that higher LRP1 protein production might be linked to increased endocytosis of upregulated transporter proteins at the cell surface, resulting in enhanced accumulation of dox and emodin and subsequent growth inhibition. This suggests that elevating LRP1 expression could be a promising target for interventions to improve the efficacy of anticancer drugs. PMID: 29644529
  3. This study is the first to demonstrate that the sLR11 gene rs3824968 polymorphism of factor T may increase the risk of carotid atherosclerosis in Type 2 Diabetes Mellitus patients by regulating LDL-C concentration in Korean and Han nationalities in China. PMID: 29865095
  4. A common polymorphism reduces LRP1 mRNA stability and is associated with elevated plasma factor VIII levels. PMID: 28431990
  5. Circulating sLRP1 is a novel biomarker that correlates with EAT volume in T1DM patients. PMID: 29348672
  6. Data suggests that amyloid precursor protein (APP) dimerization influences its interaction with LDL receptor-related protein 1 (LRP1) and LDL-receptor related protein SorLA (SorLA), implying that APP dimerization modulates its interplay with sorting molecules, ultimately affecting its localization and processing. PMID: 28799085
  7. LRP1 facilitates cell adhesion by coordinating a multi-protein pathway that activates, traffics, and degrades integrins. This suggests that LRP1 acts as a crucial component in the integrin quality control system, ensuring a robust connection to the extracellular matrix. PMID: 29116364
  8. This study identifies LRP1 as a regulator of CXCR3, which could have significant implications for tumor biology. PMID: 29146996
  9. The C4408R mutant, located at the APP695 alpha-secretase cleavage site of LRP1, when expressed in CHO cells expressing APPswe or wild-type APP (APPwt), co-expression of LRP1-CT C4408R decreases Abeta and increases sAPPalpha and alpha-CTF compared with co-expression of wild-type LRP1-CT. LRP1-CT C4408R enhances the unglycosylated form of LRP1-CT and reduces APP endocytosis. PMID: 28612181
  10. In cerebral blood vessels, LRP1 is a key mediator for the rapid removal of ABETA from the brain via transport across the blood-brain barrier. This research summarizes recent findings on LRP1 function and discusses the targeting of LRP1 as a modulator for Alzheimer's Disease pathology and drug delivery into the brain. [review] PMID: 28948494
  11. Extracellular vesicles do not contribute to higher circulating levels of soluble LRP1 in idiopathic dilated cardiomyopathy. PMID: 28557183
  12. LRP-1 silencing leads to a reduction in cell migratory capacity in a 3D configuration. PMID: 27463962
  13. LRP1 plays a role in insulin signaling and potentially acts as a link between lipoprotein and glucose metabolism in diabetes. [review] PMID: 28584820
  14. Findings indicate that holo-Lf, but not apo-Lf, increases TE expression through LRP-1 in human dermal fibroblasts. This suggests that holo-Lf and TGF-beta1 enhance TE expression by activating the PI3K/Akt1 and PI3K/Akt2 pathways, respectively. PMID: 28833753
  15. Development of a monoclonal anti-ADAMTS-5 antibody that specifically blocks its interaction with LRP1 has been reported. PMID: 28306378
  16. MMP-13 might be involved in the physiological turnover of cartilage extracellular matrix, and LRP1 is a key modulator of extracellular levels of MMP-13. Its internalization is independent of ADAMTS-4, -5, and TIMP-3 levels. PMID: 27084377
  17. Research has investigated the interaction between TIMP3 and LRP1 using a synthetic analog of the LRP1 receptor. PMID: 27476612
  18. FVIIa-antithrombin, but not FVIIa, is a ligand for LRP1. LRP1 contributes to the clearance of FVIIa-antithrombin in vivo. PMID: 27614059
  19. Activated alpha2 -Macroglobulin Induces Mesenchymal Cellular Migration Of Raw264.7 Cells Through Low-Density Lipoprotein Receptor-Related Protein 1. PMID: 28012205
  20. A study demonstrated that LRP1 expression is significantly upregulated by myeloid cells in active multiple sclerosis lesions compared to surrounding healthy tissue. Findings suggest that LRP1 functions in microglia to maintain an anti-inflammatory and neuroprotective state during inflammatory insult. PMID: 27400748
  21. Low LRP1 expression in T cells is attributed to shedding. Integrin ligands and CXCL12 antagonize shedding through a TSP-1-dependent pathway, and ligation of CD28 antagonizes shedding independently of TSP-1. PMID: 28580688
  22. Altered Met receptor phosphorylation and LRP1-mediated uptake occur in cells lacking carbohydrate-dependent lysosomal targeting. PMID: 28724630
  23. LRP1 single-nucleotide polymorphism is associated with migraine. PMID: 27322543
  24. Electrostatic potential calculations suggest a competition between negatively charged GAGs and highly negatively charged complement-like domains of LRP-1 for binding to a positively charged area of TIMP-3 as an underlying mechanism. PMID: 27610455
  25. Research revealed a pH-dependent release of the ligand associated with a conformational change of the receptor. This comprehensive investigation of the complete LRP1 ectodomain significantly advances our understanding of this vital receptor and provides a foundation for further elucidating the mechanism of action of LRP1 in a whole and integrated system. PMID: 27956551
  26. While fVIII bound avidly to soluble forms of clusters II and IV from LRP1, only soluble cluster IV competed with the binding of fVIII to full-length LRP1, revealing that cluster IV represents the primary fVIII binding site in LRP1. PMID: 27794518
  27. LRP1 protein expression in human abdominal aortic aneurysm tissues may be downregulated by miR-205 through translational inhibition, leading to reduced clearance of pericellular MMP-9 and promoting aneurysm formation. PMID: 26781079
  28. Data indicates that D1D2 is capable of binding to a distinct site on LRP1 to form a monovalent complex. These studies confirm the canonical model for ligand recognition by this class of receptors, which is initiated by pairs of lysine residues that dock into acidic pockets on the receptor. PMID: 27402839
  29. Results suggest that genetic variations in LRP1 and ULK4 contribute to the risk of presenting with an acute aortic dissection. PMID: 27569546
  30. Considering that LRP-1, by mediating the clearance of matrix metalloproteinases, is involved in the regulation of extracellular matrix remodeling and cell migration, we conclude that decreased expression of LRP-1 could contribute to the increased activity of MMPs observed in cancers. PMID: 27931798
  31. These data indicate that the Rho/ROCK signaling pathway is involved in HIV-1 Tat-mediated changes in occludin, RAGE, and LRP1 in human cerebral microvascular endothelial cells. PMID: 27563375
  32. Studies demonstrate a role for ADAM10 in the ectodomain shedding of LRP1 in the brain and the clearance of Abeta across the blood-brain barrier. This could provide a novel strategy for attenuating Abeta accumulation in the AD brain. PMID: 27503326
  33. Research shows that TTR acts as a carrier of Abeta at the blood-brain-barrier and liver, utilizing LRP1. PMID: 26837706
  34. Studies highlight LRP1's ability to promote endocytosis and deliver cell signaling, suggesting multiple roles in tumorigenesis and tumor progression. Additionally, LRP1 exhibits dual effects on tumor cell invasion and migration. Furthermore, LRP1 can be regulated through methylation of its CpG islands. [review] PMID: 26738504
  35. A study demonstrated that 1,25(OH)2D3, the active form of vitamin D, plays a crucial role in increasing Abeta1-40 vectorial transport from the brain to blood and systemic clearance from peripheral circulation. It achieves this by increasing LRP1 levels both in vivo and in vitro, and reducing RAGE level in the blood-brain barrier model in vitro. PMID: 26820600
  36. Circulating soluble LRP1 concentrations were associated with the occurrence of carotid atherosclerosis in a hypercholesterolemia population. PMID: 26285183
  37. LRP1 is identified as a pathogenic gene for autosomal recessive Keratosis pilaris atrophicans and keratosis pilaris. PMID: 26142438
  38. Research suggests a role for LRP1/PARP1 signaling in endothelial cell proliferation and retinal neovascularization induced by hypoxia. PMID: 26634655
  39. A cell surface-expressed beta-galactoside binding protein, galectin-8, was suggested to be involved in factor V endocytosis. PMID: 25800007
  40. alpha1-antitrypsin internalization is a clathrin-dependent and low-density lipoprotein receptor-related protein 1-mediated endocytosis process. PMID: 26206901
  41. LRP1, p38 MAPK, and ERK1/2 mediate the internalization of beta-amyloid 1-42 in neurons. PMID: 25936756
  42. In conclusion, the C667T polymorphism of LRP-1 is moderately but significantly associated with global and regional amyloid deposition in AD. PMID: 24596678
  43. LRP1 variants may be associated with the risk of ischemic stroke. PMID: 26031789
  44. Lrp1-antisense directly binds to high-mobility group box 2 (Hmgb2) and inhibits the activity of Hmgb2 to enhance Srebp1a-dependent transcription of Lrp1. PMID: 25937287
  45. Statistically significant associations were also found for the PS1 1/2 polymorphism in both the dominant and recessive genetic models, whereas no association was found for the LRP C/T polymorphism. PMID: 25730041
  46. Data suggests that aortic but not circulating LRP1 is downregulated in patients with AAA and indicates a potential role for this protein in clearing an aneurysm-relevant ligand. PMID: 26188720
  47. Two recurrent fusion genes associated with the 12q locus, LRP1-SNRNP25 and KCNMB4-CCND3, were identified by RT-PCR, Sanger sequencing, and FISH. These genes were found to be osteosarcoma specific in a validation cohort of 240 other sarcomas. PMID: 25300797
  48. The interaction between factor VIII and LRP1 occurs over an extended surface containing multiple lysine residues. PMID: 25903134
  49. Domain CR9 appears to be critical for LRP1-mediated AgLDL binding and internalization in hVSMCs. These findings open new avenues for an innovative anti-VSMC foam cell-based strategy for the treatment of vascular lipid deposition in atherosclerosis. PMID: 25918169
  50. The rs1466535 LRP1 polymorphism is not a significant and independent risk factor for carotid artery stenosis. PMID: 25238221

Show More

Hide All

Database Links

HGNC: 6692

OMIM: 107770

KEGG: hsa:4035

STRING: 9606.ENSP00000243077

UniGene: Hs.162757

Involvement In Disease
Keratosis pilaris atrophicans (KPA)
Protein Families
LDLR family
Subcellular Location
[Low-density lipoprotein receptor-related protein 1 85 kDa subunit]: Cell membrane; Single-pass type I membrane protein. Membrane, coated pit.; [Low-density lipoprotein receptor-related protein 1 515 kDa subunit]: Cell membrane; Peripheral membrane protein; Extracellular side. Membrane, coated pit.; [Low-density lipoprotein receptor-related protein 1 intracellular domain]: Cytoplasm. Nucleus.; Golgi outpost. Cytoplasm, cytoskeleton, microtubule organizing center.
Tissue Specificity
Most abundant in liver, brain and lung.

Q&A

What is LRP1 and why is it important in biomedical research?

LRP1 (Low-density lipoprotein receptor-related protein 1) is a large endocytic receptor involved in multiple cellular processes including intracellular signaling, lipid homeostasis, and clearance of apoptotic cells . It functions as both a scavenger and signaling molecule, binding and internalizing numerous plasma components . Most notably, LRP1 is necessary for alpha-2-macroglobulin (A2M)-mediated clearance of secreted amyloid precursor protein and beta-amyloid, the main component of amyloid plaques found in Alzheimer's disease patients . Expression of this gene decreases with age and has been found to be lower in brain tissue from Alzheimer's patients, highlighting its potential role in neurodegeneration . Additionally, LRP1 dysfunction has been linked to developmental dysplasia of the hip, osteoporosis, and osteoarthritis, making it a critical target for musculoskeletal research .

What are the key advantages of using recombinant rabbit monoclonal antibodies for LRP1 detection?

Recombinant rabbit monoclonal antibodies offer several significant advantages over traditional antibodies for LRP1 detection. These antibodies are produced using in vitro expression systems by cloning specific antibody DNA sequences from immunoreactive rabbits, followed by screening individual clones to select optimal candidates for production . This approach results in better specificity and sensitivity compared to conventional antibodies, ensuring more reliable experimental outcomes in LRP1 research . Additionally, these antibodies provide excellent lot-to-lot consistency, which is crucial for longitudinal studies and reproducible research . The animal origin-free formulations reduce potential contamination concerns, while the broader immunoreactivity to diverse targets leverages the larger rabbit immune repertoire, enabling detection of various LRP1 epitopes and isoforms . These technical improvements make recombinant rabbit monoclonal antibodies particularly valuable for complex proteins like LRP1, which has multiple domains and undergoes post-translational modifications.

What experimental applications are suitable for LRP1 recombinant monoclonal antibodies?

LRP1 recombinant monoclonal antibodies are versatile research tools applicable across multiple experimental platforms. They can be effectively utilized in flow cytometry for quantitative assessment of LRP1 expression on cell surfaces, as demonstrated in studies using HEK293-EBNA1 cells expressing LRP1-GFP . Immunocytochemistry and immunohistochemistry applications allow for visualization of LRP1 distribution in cultured cells and tissue sections, respectively, revealing both surface expression and subcellular localization in compartments such as endosomes . For protein interaction studies, these antibodies perform well in immunoprecipitation assays, enabling isolation of LRP1 and its binding partners . Western blot applications provide quantitative assessment of LRP1 protein levels and processing, which is particularly important given that LRP1 undergoes significant post-translational modifications including glycosylation, protein cleavage, and phosphorylation . Each application requires specific optimization, including appropriate blocking agents, incubation times, and detection systems to maximize signal-to-noise ratio.

What is known about LRP1 structure and how does this influence antibody selection?

LRP1 presents unique structural challenges that directly impact antibody selection strategies. As the largest known mammalian endocytic receptor, LRP1 is highly modular with a complex arrangement of 61 domains in its full-length ectodomain . The protein is extensively glycosylated and cysteine-rich, making recombinant expression and antibody production technically challenging . The human canonical protein has a reported length of 4544 amino acid residues and a molecular mass of approximately 504.6 kDa, with up to two different isoforms identified . When selecting antibodies, researchers should consider the specific epitope location, as some antibodies target synthetic peptides within human LRP1 amino acids 4,471-4,520, which may affect detection of specific domains or processed forms of the protein . Additionally, antibodies should be validated for cross-reactivity with LRP1 orthologs from relevant experimental models, as LRP1 gene orthologs have been reported in mouse, rat, bovine, frog, chimpanzee, and chicken species . Understanding these structural complexities ensures selection of appropriate antibodies for specific experimental questions.

How can I optimize immunodetection protocols for LRP1 in tissues with variable expression levels?

Optimizing immunodetection protocols for LRP1 requires a strategic approach to address its variable expression across different tissues. Begin by conducting a thorough antigen retrieval optimization, testing both heat-induced epitope retrieval (HIER) methods using citrate buffer (pH 6.0) and Tris-EDTA buffer (pH 9.0), as well as enzymatic retrieval using proteinase K, particularly for formalin-fixed tissues . The buffer composition should be adjusted based on the target tissue, with TBS containing 0.05% BSA recommended for maintaining antibody stability and reducing non-specific binding . Given LRP1's notable expression in liver, brain, and lung tissues, signal amplification techniques may be necessary for tissues with lower expression levels . For dual or multi-color immunostaining, sequential staining protocols are preferable to simultaneous approaches to minimize cross-reactivity, particularly when detecting LRP1 alongside Wnt5a due to their partial colocalization in developing tissues . Include positive control tissues with confirmed high LRP1 expression (such as liver sections) and negative controls (primary antibody omission or isotype controls) in each experimental run to validate staining specificity and establish detection thresholds.

What strategies exist for investigating LRP1-mediated endocytosis in live cell imaging experiments?

Investigation of LRP1-mediated endocytosis through live cell imaging requires sophisticated methodological approaches to capture this dynamic process. Begin by generating stable cell lines expressing fluorescently tagged LRP1 constructs, such as LRP1-GFP, which has been successfully expressed in HEK293-EBNA1 cells and visualized on cell surfaces and in subcellular compartments . Complement this with fluorescently labeled ligands known to bind LRP1, such as receptor-associated protein (RAP), to track the binding, internalization, and intracellular trafficking in real-time . For optimal visualization, spinning disk confocal microscopy with environmental control chambers maintaining 37°C and 5% CO2 is recommended to maintain physiological endocytic rates. Implement pulse-chase experiments using pH-sensitive fluorophores like pHrodo™ to distinguish between surface-bound and internalized ligands, while quantifying endocytic rates through automated tracking algorithms measuring fluorescence intensity changes over time. To investigate the role of specific endocytic machinery components, combine imaging with siRNA-mediated knockdown of clathrin, caveolin, or other endocytic adaptors, or pharmacological inhibitors like dynasore (dynamin inhibitor) or chlorpromazine (clathrin inhibitor). This multifaceted approach enables comprehensive analysis of LRP1's endocytic function in diverse cellular contexts.

How can I explore the interactions between LRP1 and Wnt signaling pathways in developmental research?

Investigating interactions between LRP1 and Wnt signaling pathways requires a multidisciplinary experimental design integrating molecular, cellular, and developmental approaches. First, utilize co-immunoprecipitation with LRP1 recombinant monoclonal antibodies to confirm direct binding interactions between LRP1 and Wnt5a in your specific experimental model, as this interaction has been demonstrated to regulate non-canonical Wnt/planar cell polarity (PCP) components . Complement biochemical approaches with proximity ligation assays (PLA) to visualize and quantify these interactions in situ within developing tissues. For functional analysis, develop conditional knockout models with tissue-specific deletion of LRP1 in skeletal progenitors, which has been shown to cause joint fusion, malformation of cartilage/bone templates, and delayed primary ossification . Monitor alterations in abundance and distribution of core PCP components like Wnt5a and Vangl2 through immunofluorescence microscopy and quantitative image analysis . Implement TOPFlash/FOPFlash reporter assays to measure effects on canonical Wnt signaling, while simultaneously assessing PCP pathway activation through JNK phosphorylation status. Cross-validate findings through morpholino-mediated knockdown experiments in Xenopus models, which provide a tractable system for investigating LRP1's regulatory role in Wnt/PCP signaling during embryonic development .

What are the key considerations when using LRP1 antibodies for studying neurodegenerative diseases?

When employing LRP1 antibodies in neurodegenerative disease research, several critical factors must be considered to ensure robust and interpretable results. First, antibody selection should account for age-dependent decreases in LRP1 expression, which is particularly relevant for Alzheimer's disease studies where LRP1 levels are lower than controls in patient brain tissue . Sample preparation protocols must preserve LRP1's integrity while enabling detection within the complex neural architecture; this typically requires optimization of fixation parameters (4% paraformaldehyde for 24-48 hours) and cryoprotection procedures for frozen sections. When conducting comparative analyses between diseased and healthy tissues, implement quantitative immunofluorescence with standardized image acquisition settings and calibration standards to account for background autofluorescence, which is often elevated in neurodegenerative tissues. Co-staining with markers for amyloid plaques, neurofibrillary tangles, or neuronal/glial markers provides contextual information about LRP1's relationship to pathological features . For functional studies investigating LRP1's role in A2M-mediated clearance of amyloid precursor protein and beta-amyloid, combine immunodetection with biochemical clearance assays using fluorescently labeled substrates. These methodological refinements enhance the sensitivity and specificity of LRP1 detection in neurodegenerative disease contexts.

How can I address weak or inconsistent signal issues when using LRP1 recombinant monoclonal antibodies?

Weak or inconsistent signals when using LRP1 recombinant monoclonal antibodies often stem from multiple technical factors that can be systematically addressed. Begin by verifying antibody concentration and storage conditions, as the recommended working concentration of 1 mg/mL may require titration for specific applications, and aliquoting stock solutions prevents freeze-thaw cycles that diminish activity . Next, optimize antigen retrieval methods, as LRP1's complex structure with extensive glycosylation and numerous disulfide bonds may obscure epitopes; test extended retrieval times or combinatorial approaches using both heat and enzymatic methods. Consider signal amplification strategies using tyramide signal amplification (TSA) or polymer-based detection systems, which can enhance sensitivity without increasing background. Evaluate fixation protocols, as overfixation can mask epitopes while underfixation may compromise tissue architecture; for cell lines, 4% paraformaldehyde for 10-15 minutes is typically optimal, while tissues may require longer fixation with careful monitoring. If inconsistency persists, implement a dual antibody approach using two different monoclonal antibodies targeting distinct LRP1 epitopes to confirm staining patterns. Finally, verify expression levels through complementary techniques such as qRT-PCR, as LRP1 expression varies considerably across tissues and developmental stages.

What approaches help distinguish between different LRP1 isoforms or processed forms?

Distinguishing between LRP1 isoforms or processed forms requires a strategic experimental design that leverages both antibody properties and complementary techniques. First, select epitope-specific antibodies that target distinct regions of the protein; for instance, antibodies recognizing the N-terminal region versus those binding the C-terminal intracellular domain can differentiate between full-length LRP1 and its processed fragments . Implement gradient gel electrophoresis (3-8% Tris-Acetate gels) for Western blotting to achieve better separation of high molecular weight proteins, enabling visualization of the ~515 kDa full-length LRP1 versus the ~85 kDa light chain that results from furin-mediated cleavage. Combine immunoprecipitation with mass spectrometry to identify specific post-translational modifications, including glycosylation patterns and phosphorylation sites that may differ between isoforms . For in situ analyses, utilize RNAscope® technology with isoform-specific probes to determine which transcript variants are expressed in specific tissues or cell types. Finally, conduct pulse-chase experiments with metabolic labeling to track the processing and turnover rates of different LRP1 forms, providing insights into the dynamic regulation of this complex receptor under various experimental conditions.

How can potential cross-reactivity with other LDLR family members be assessed and minimized?

Assessing and minimizing cross-reactivity with other low-density lipoprotein receptor (LDLR) family members requires rigorous validation strategies due to structural homology within this protein family. Begin by conducting comprehensive sequence alignment analyses comparing the immunogen sequence (such as the synthetic peptide within Human LRP1 aa 4,471-4,520) against all LDLR family members to identify regions of homology that might promote cross-reactivity . Implement competitive binding assays using recombinant proteins representing different LDLR family members to determine binding specificity quantitatively. For cellular systems, utilize CRISPR/Cas9-mediated knockout of LRP1 as the gold standard negative control to confirm antibody specificity, while systematically overexpressing individual LDLR family members to detect potential cross-reactivity. When cross-reactivity is identified, employ pre-adsorption protocols by incubating the antibody with the recombinant protein containing the cross-reactive epitope prior to the primary application. For immunohistochemistry applications, include tissues from LRP1-knockout models as negative controls and compare staining patterns with established expression profiles of other LDLR family members. Finally, validate key findings with multiple antibody clones recognizing different LRP1 epitopes to confirm that observed signals truly represent LRP1 rather than related receptors.

What are the emerging applications of LRP1 antibodies in studying skeletal development and pathologies?

LRP1 antibodies are increasingly employed in cutting-edge skeletal research, revealing novel insights into development and disease mechanisms. Recent studies demonstrate abundant LRP1 expression in skeletal progenitor cells beginning at mouse embryonic stage E10.5, particularly in the perichondrium, the critical stem cell layer surrounding developing limbs that is essential for bone formation . Implementing lineage-specific conditional knockout models with subsequent immunostaining has revealed that LRP1 deficiency in skeletal stem cells causes profound developmental abnormalities, including joint fusion, malformation of cartilage/bone templates, and markedly delayed or absent primary ossification . These phenotypes manifest as severe skeletal defects including hip joint and patella deficiencies, deformed low-density long bones, dwarfism, and impaired mobility . Using co-immunoprecipitation with LRP1 antibodies has demonstrated the receptor's direct interaction with Wnt5a, establishing a mechanistic link between LRP1 and the non-canonical Wnt/planar cell polarity pathway critical for proper skeletal patterning . Combined immunohistochemistry and genetic approaches have implicated LRP1 dysfunction in developmental dysplasia of the hip, osteoporosis, and osteoarthritis, suggesting potential diagnostic applications for LRP1 antibodies in identifying patients at risk for these conditions .

How can LRP1 recombinant monoclonal antibodies be utilized in therapeutic development research?

LRP1 recombinant monoclonal antibodies offer powerful tools for therapeutic development research across multiple disease contexts. For Alzheimer's disease interventions, these antibodies enable screening of compounds that enhance LRP1-mediated clearance of beta-amyloid, capitalizing on LRP1's role in A2M-mediated clearance of amyloid precursor protein and beta-amyloid . High-content imaging platforms utilizing fluorescently labeled antibodies can quantify LRP1 surface expression changes in response to drug candidates, providing a functional readout for therapeutic efficacy. In osteoarthritis research, antibody-based blocking strategies targeting specific LRP1 domains can help elucidate which interactions might be therapeutically modified to prevent cartilage degradation or stimulate regeneration . For targeted drug delivery systems, conjugating therapeutic agents to anti-LRP1 antibody fragments or mimetic peptides exploits LRP1's endocytic capacity to enhance cellular uptake in tissues with high LRP1 expression, such as the liver, brain, and lung . Flow cytometry with LRP1 antibodies facilitates patient stratification in clinical trials by quantifying receptor levels in accessible tissues, potentially identifying individuals most likely to respond to LRP1-targeted therapies. These diverse applications highlight the translational value of LRP1 recombinant monoclonal antibodies in bridging basic research and therapeutic development.

What methodological advances are enabling structural studies of LRP1 using antibody-based approaches?

Recent methodological breakthroughs have revolutionized structural studies of LRP1 using antibody-based approaches, overcoming the historical challenges of working with this large, highly glycosylated, and cysteine-rich protein. The development of multistep cloning approaches coupled with DNA dilution techniques has enabled the first successful recombinant expression of the complete 61 domains of the full-length LRP1 ectodomain, providing unprecedented opportunities for structural characterization . Single-particle cryo-electron microscopy combined with Fab fragments from recombinant monoclonal antibodies now permits visualization of LRP1's three-dimensional organization in both ligand-bound and unbound states. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) paired with epitope-specific antibodies facilitates mapping of conformational changes upon ligand binding, particularly valuable for understanding LRP1's interaction with receptor-associated protein (RAP) . Surface plasmon resonance (SPR) using immobilized antibodies provides quantitative binding kinetics for LRP1-ligand interactions under varying pH conditions, critical for elucidating the receptor's endocytic mechanism. X-ray crystallography of LRP1 fragments complexed with antibody fragments has begun to reveal domain-specific structures at atomic resolution. Together, these complementary approaches are generating an integrated structural understanding of this complex receptor that was previously unattainable due to technical limitations.

How should quantitative data from LRP1 immunodetection be normalized and statistically analyzed?

Robust normalization and statistical analysis of quantitative LRP1 immunodetection data requires careful consideration of biological and technical variables. For Western blot quantification, normalize LRP1 band intensities to appropriate loading controls based on cellular compartment; use GAPDH or β-actin for cytoplasmic fractions, Na+/K+ ATPase for membrane fractions, and Lamin A/C for nuclear extracts . When analyzing flow cytometry data, implement fluorescence minus one (FMO) controls to establish proper gating strategies, and express results as median fluorescence intensity (MFI) rather than percent positive cells to capture the full spectrum of expression levels . For immunohistochemistry quantification, normalize signal intensity to tissue area or cell number, and employ multiple randomly selected fields (minimum 5-10 per sample) to account for heterogeneous expression patterns. Account for batch effects through randomized processing and analysis of samples across experimental groups, and consider ANCOVA models with batch as a covariate when combining data from multiple experimental runs. For developmental studies tracking LRP1 expression changes, pair-wise statistical comparisons should be performed using appropriate tests based on data distribution (parametric vs. non-parametric), with correction for multiple comparisons (e.g., Bonferroni or Benjamini-Hochberg procedures). Finally, report effect sizes alongside p-values to convey the magnitude of observed differences in addition to statistical significance.

What approaches help resolve contradictory findings when using different LRP1 antibody clones?

Contradictory findings when using different LRP1 antibody clones can be systematically resolved through a structured investigative approach. Begin by conducting a detailed epitope mapping analysis to determine precisely where each antibody binds within LRP1's complex structure, as epitope accessibility may vary depending on protein conformation, post-translational modifications, or interaction with binding partners . Implement a multi-antibody validation panel testing at least three independent antibody clones targeting distinct epitopes under identical experimental conditions to identify consensus patterns versus clone-specific anomalies. Evaluate each antibody's specificity using tissues or cells from LRP1 knockout models as negative controls, and confirm binding to recombinant LRP1 protein through ELISA or surface plasmon resonance to quantify affinity and specificity parameters. Consider the influence of sample preparation methods, as different fixation protocols, antigen retrieval methods, or buffer compositions may differentially affect epitope accessibility for each antibody clone . Complement antibody-based detection with orthogonal techniques such as RNA-seq, mass spectrometry, or functional assays to corroborate protein expression and activity findings. Finally, consult literature reports of similar contradictions and incorporate knowledge of LRP1's tissue-specific processing and interactions to develop a unified model that accommodates seemingly discrepant observations.

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.