Phospho-L1CAM (S1181) Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
The antibody is supplied in a liquid form, buffered in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide.
Form
Liquid
Lead Time
Generally, we can ship your order within 1-3 business days after receiving it. Delivery time may vary depending on the purchase method and location. Please consult your local distributor for specific delivery time details.
Synonyms
Antigen identified by monoclonal antibody R1 antibody; CAML1 antibody; CD171 antibody; CD171 antigen antibody; HSAS antibody; HSAS1 antibody; Hyd antibody; L1 antibody; L1 cell adhesion molecule antibody; L1-NCAM antibody; L1cam antibody; L1CAM_HUMAN antibody; MASA antibody; MIC5 antibody; N CAML1 antibody; N-CAM-L1 antibody; NCAM-L1 antibody; NCAML1 antibody; Nerve-growth factor-inducible large external glycoprotein antibody; Neural cell adhesion molecule L1 antibody; NILE antibody; OTTHUMP00000025992 antibody; S10 antibody; SPG1 antibody
Target Names
Uniprot No.

Target Background

Function
L1CAM, also known as Neural cell adhesion molecule, is a transmembrane protein involved in regulating cell adhesion dynamics and generating transmembrane signals at tyrosine kinase receptors. During brain development, L1CAM plays a crucial role in several processes, including neuronal migration, axonal growth and fasciculation, and synaptogenesis. In the mature brain, L1CAM continues to play a significant role in the dynamics of neuronal structure and function, including synaptic plasticity.
Gene References Into Functions
  1. In uterine carcinosarcoma, L1CAM expression was positive in the epithelial component in 65.4% of cases, while it was negative in the mesenchymal component. Notably, in cases with intermingled components, expression was confirmed to be limited to the epithelial component through a double stain for L1CAM and keratin. The expression of L1CAM did not correlate with overall or disease-free survival. PMID: 30140948
  2. Our findings suggest that L1CAM may be involved in the pathogenesis of at least a subset of endometrial clear cell carcinomas. PMID: 28941294
  3. The directional force for laminin-induced growth cone haptotaxis is generated by the grip and slip of L1-CAM on the substrates, which occur asymmetrically under the growth cone. PMID: 29483251
  4. L1CAM promotes esophageal squamous cell carcinoma tumorigenicity by upregulating ezrin expression. PMID: 28939985
  5. TWIST1, potentially via GAS6 and L1CAM, led to higher expression and activation of Akt upon cisplatin treatment. Notably, inhibition of Akt activation sensitized cells to cisplatin. PMID: 27876874
  6. The data suggests that L1CAM is a highly interesting therapeutic target to prevent further metastatic spread in melanoma patients. PMID: 29432466
  7. High circulating levels of autoantibodies against L1-cell adhesion molecule are associated with esophageal squamous cell carcinoma. PMID: 28181176
  8. A functional role for L1CAM in extrahepatic cholangiocarcinoma carrying the activating KRAS mutation has been observed. L1CAM promotes cell migration and invasion via JNK activation in extrahepatic cholangiocarcinoma. PMID: 28535665
  9. This review and meta-analysis concludes that L1CAM might be an effective poor prognostic factor for patients with various tumor types. PMID: 27833079
  10. High L1CAM expression is associated with vulvar squamous cell carcinomas. PMID: 27028855
  11. Our preclinical assessment of the CE7 epitope on CD171 supports its utility and safety as a CAR T-cell target for neuroblastoma immunotherapy. PMID: 27390347
  12. L1CAM may have a role in human endometrial cancer, and miR-34a has an inverse relationship to L1CAMEXP. PMID: 27233077
  13. L1CAM mRNA expression appears to play a significant role in the pathophysiology of ovarian cancer, which is translated into poor clinical outcome. PMID: 27174921
  14. These results suggest that a deficiency in L1 may partially account for Rett syndrome phenotypes. PMID: 29050935
  15. L1CAM failed to be a clinically relevant marker of poor prognosis in stage I endometrioid endometrial carcinoma. PMID: 27488577
  16. This study revealed an unexpected role of L1CAM in the pathological crosstalk between the immune and nervous systems. PMID: 27544757
  17. Mutations involving L1 cell adhesion molecule are associated with chemotherapy-resistant urothelial carcinoma. PMID: 27749842
  18. Data suggest that targeted therapy to neural cell adhesion molecule L1 (L1) might be effective in the treatment of retinoblastoma tumors. PMID: 28061460
  19. CD10 is a necessary component conferring the L1 effects in CRC cells. The identification of gene expression patterns of L1-domain-specific point mutations may provide novel markers and targets for interfering with L1-mediated CRC progression. PMID: 27641335
  20. High L1-CAM expression is associated with low radiosensitivity in neuroblastoma. PMID: 27432152
  21. Neural cell adhesion molecule L1 (L1CAM)-mediated cell-cell aggregation was severely impaired by L1CAM variants p.I37N, p.M172I and p.D202Y, but was preserved by the variant p.T38M. PMID: 26891472
  22. The differential expression timing of CD184 and CD171 permits identification and enrichment of RGCs from retinal organoids at differing maturation states from committed progenitors to differentiating neurons. PMID: 27867005
  23. This study examined the spatiotemporal distribution of L1CAM in the early human fetal period using immunohistochemistry and in situ hybridization. In advanced differentiated epithelia, such as those of the gastrointestinal system, L1CAM localization vanishes. However, in epithelia undergoing further development, such as those of the urogenital system, L1CAM is further needed for their complete establishment. PMID: 28026654
  24. The expression of L1CAM was associated with advanced stage, nodal involvement, high tumor grade, non-endometrioid histology, lymphovascular space invasion, and distant recurrences in all cases, and with reduced survival in endometrial carcinomas, but not in the non-endometrioid carcinomas. L1CAM may induce EMT-like changes, but seems to only play a role in metastasis, not in invasion. PMID: 27505134
  25. L1CAM expression is an independent predictor of poor survival in endometrial cancer and is associated with advanced stage, high-risk endometrial cancer. PMID: 26861585
  26. L1CAM is a neuronal cell adhesion molecule involved in the development of the nervous system and progression of malignancies. (Review) PMID: 27267927
  27. Splicing variant c.1267+5delG was identified in fetal hydrocephalus. The same mutation and severe L1 syndrome were confirmed in the second pregnancy. PMID: 27207492
  28. Involvement of L1CAM in the regulation of activity of the canonical Wnt pathway and expression of genes of class I melanoma-associated antigens in melanoma. PMID: 27165065
  29. L1CAM was a significant independent prognosticator for disease-specific survival in endometrial carcinoma. PMID: 27695947
  30. Reports show a high frequency of L1CAM expression in high-risk endometrial cancers associated with mutant p53 expression. PMID: 26743472
  31. L1CAM is frequently expressed in testicular germ cell tumors but not in normal testis. PMID: 26933044
  32. L1 syndrome should be considered in the differential diagnosis of intellectual disability or mental retardation in children, especially when other signs such as hydrocephalus or adducted thumbs are present. PMID: 25948108
  33. Genes induced during L1-mediated colorectal cancer cell metastasis. PMID: 26399194
  34. Our results suggest that the overexpression of L1CAM may be related to several established markers of poor prognosis in breast cancer patients. PMID: 26464672
  35. L1-CAM and N-CAM: From Adhesion Proteins to Pharmacological Targets. PMID: 26478212
  36. The CE7-epitope of L1-CAM is a cell adhesion molecule aberrantly expressed in several cancers and may have a role in immunotherapy. PMID: 26761817
  37. Novel missense variant in L1CAM was identified in two Caucasian families with mild-moderate intellectual disability without obvious L1 syndrome features. PMID: 25934484
  38. This study identified predicted pathogenic, hemizygous variants on chromosome X in disease genes L1CAM. PMID: 25666757
  39. Our findings establish Slug-induced L1CAM expression as a mediator of a chemoresistant and migratory phenotype in pancreatic adenocarcinoma cells. PMID: 25860483
  40. The expression level of L1CAM were negatively correlated with miR-503 levels in osteosarcoma tissues. PMID: 25536034
  41. L1CAM is expressed in triple-negative breast cancers and is inversely correlated with androgen receptor. PMID: 25510351
  42. A positive relationship between L1 and pPKD1 in both cultured cerebellar neurons and human cerebellar tissue suggests that L1 functions in the modulation of PKD1 phosphorylation. PMID: 25445362
  43. This indicates that similar biofunctionalization approaches based on N-cadherin and L1 can be translated to 3-D "transplantable" scaffolds with enhanced neurotrophic behaviors. PMID: 24914828
  44. Findings shed new light on the complex regulation of L1CAM in cancers and advocate the use of L1CAM/miR-21-3p for diagnostic application. PMID: 25149066
  45. The data show that L1CAM promotes the enrichment of immunosuppressive T cells, particularly of a CD4(+)CD25(-)CD69(+)-phenotype in pancreatic ductal adenocarcinoma, providing a novel mechanism of tumor immune escape that contributes to tumor progression. PMID: 24746181
  46. Data indicate that positive L1 cell adhesion molecule (L1CAM) expression was significantly correlated with the risk of distant recurrence. PMID: 25126672
  47. Overexpression of L1CAM is associated with tumor progression via ERK signaling in gastric cancer. PMID: 24046108
  48. Expression of L1CAM and EPCAM in gastric cancer was significantly associated with lymph node and distant metastasis, and poor prognosis. PMID: 24422715
  49. Human pathological H210Q, R184Q, and Y1070C, but not the E309K and L120V L1CAM mutations, affect outside-in signaling via the FIGQY Ankyrin binding domain, which is required for synapse formation. PMID: 24155914

Show More

Hide All

Database Links

HGNC: 6470

OMIM: 303350

KEGG: hsa:3897

STRING: 9606.ENSP00000359074

UniGene: Hs.522818

Involvement In Disease
Hydrocephalus due to stenosis of the aqueduct of Sylvius (HSAS); Mental retardation, aphasia, shuffling gait, and adducted thumbs syndrome (MASA); Agenesis of the corpus callosum, X-linked, partial (ACCPX)
Protein Families
Immunoglobulin superfamily, L1/neurofascin/NgCAM family
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Cell projection, growth cone. Cell projection, axon. Cell projection, dendrite.

Q&A

What is the molecular basis for L1CAM S1181 phosphorylation and its functional significance?

L1CAM (L1 Cell Adhesion Molecule) is a neuronal cell adhesion protein with critical roles in neural development and function. Phosphorylation at serine-1181 (S1181) represents a key post-translational modification that regulates L1CAM's conformation and functional properties. Research indicates that S1181 phosphorylation is part of a sequential phosphorylation cascade that affects L1CAM signaling capacity.

Importantly, S1181 phosphorylation appears to be a prerequisite for subsequent phosphorylation at threonine-1172 (T1172) by casein kinase II (CKII) . This sequential phosphorylation mechanism suggests that S1181 serves as a regulatory switch that modulates L1CAM's interactions with binding partners and influences its downstream signaling functions. The phosphorylation state at this residue likely affects the protein's three-dimensional conformation, particularly around the T1172 region, thus controlling accessibility to other kinases and binding partners.

How do Phospho-L1CAM (S1181) antibodies differ from general L1CAM antibodies in terms of epitope recognition?

Phospho-L1CAM (S1181) antibodies are specifically designed to recognize L1CAM only when phosphorylated at the serine-1181 residue, allowing researchers to distinguish between phosphorylated and non-phosphorylated forms of the protein. This specificity is achieved through immunization with synthetic phosphopeptides derived from the region surrounding S1181.

The epitope recognition of these phospho-specific antibodies differs fundamentally from general L1CAM antibodies in several ways:

FeaturePhospho-L1CAM (S1181) AntibodiesGeneral L1CAM Antibodies
ImmunogenSynthesized peptide derived from human NCAM-L1 around the phosphorylation site of S1181 Full-length or fragments of L1CAM protein
Epitope SpecificityRecognizes specifically the phosphorylated S1181 residueRecognizes various regions of L1CAM regardless of phosphorylation status
ApplicationsUsed to study phosphorylation-dependent functions and signalingUsed to study general expression and localization
Validation MethodOften validated with phosphatase treatment or phospho-null mutantsValidated with knockout/knockdown models

Understanding this distinction is crucial for experimental design when investigating phosphorylation-dependent phenomena versus general L1CAM biology.

What are the technical specifications and validation methods for commercially available Phospho-L1CAM (S1181) antibodies?

Commercially available Phospho-L1CAM (S1181) antibodies share several common specifications while differing in certain aspects. Based on the search results, these antibodies typically:

  • Are rabbit polyclonal antibodies raised against synthesized peptides derived from human NCAM-L1 around the S1181 phosphorylation site

  • React with human, mouse, and rat species

  • Are supplied in liquid form in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide

  • Can be used for Western blot (1:500-1:2000 dilution), immunohistochemistry (1:100-1:300 dilution), and ELISA (1:20000 dilution) applications

  • Require storage at -20°C or -80°C to maintain efficacy

Validation methods typically include:

  • Phospho-ELISA assays comparing reactivity with phosphorylated versus non-phosphorylated peptides

  • Western blot analysis with phosphopeptide competition/blocking to confirm specificity

  • Phosphatase treatment of samples to eliminate signal

  • Use of phospho-null mutants (S1181A) to demonstrate specificity

For optimal results, researchers should store these antibodies according to manufacturer recommendations and avoid repeated freeze-thaw cycles that could compromise antibody quality and specificity.

What are the optimized protocols for detecting Phospho-L1CAM (S1181) in different neural and tumor tissue samples?

Detection of Phospho-L1CAM (S1181) in different tissue samples requires optimized protocols tailored to specific tissue types and research questions. Here are methodological approaches for different applications:

For Immunohistochemistry (IHC):

  • Tissue Preparation: For formalin-fixed paraffin-embedded (FFPE) samples, antigen retrieval using Tris-EDTA buffer at pH 9.0 has been shown to be effective

  • Antibody Dilution: Use the antibody at 1:100-1:300 dilution (optimal concentration should be determined empirically)

  • Incubation Parameters: Incubate primary antibody at 4°C overnight

  • Signal Detection: Use secondary antibody at 1:200 dilution at room temperature for 45 minutes

  • Controls: Include phosphopeptide-blocked controls to verify specificity

For Western Blot Analysis:

  • Sample Preparation: Lyse cells or tissues in buffer containing phosphatase inhibitors to preserve phosphorylation state

  • Protein Loading: Load 20-50 μg of total protein per lane

  • Antibody Dilution: Use primary antibody at 1:500-1:2000 dilution

  • Validation Controls: Include phosphopeptide-blocked lanes to confirm specificity

  • Detection Method: Both chemiluminescence and fluorescence-based detection systems are compatible

For Neural Tissue Samples Specifically:

When working with dorsal root ganglia (DRG) or spinal cord sections, as described in the literature for studying L1CAM in neuropathic pain models, quantification should include:

  • Counting positive neurons and normalizing to the total number of DRG neurons

  • Measuring the size and number of L1CAM-immunoreactive profiles in the dorsal horn

  • Using image thresholding and quantification with software like ImageJ

These methods ensure reliable detection of phosphorylated L1CAM (S1181) across different experimental contexts.

How should researchers design experiments to investigate the relationship between L1CAM S1181 phosphorylation and TrkA receptor modulation?

Investigating the relationship between L1CAM S1181 phosphorylation and TrkA receptor modulation requires carefully designed experiments that address both the phosphorylation state of L1CAM and its interaction with TrkA signaling pathways. Based on the research showing that L1CAM positively modulates TrkA receptor activation , the following experimental approach is recommended:

1. Cell Models Selection:

  • Use high RE1-silencing transcription factor (REST) clones of PC12 cells defective in L1CAM expression

  • Consider TrkA-overexpressing cell lines for enhanced sensitivity

2. Experimental Conditions:

  • Transfect cells with wild-type L1CAM and phosphomutants (S1181A to prevent phosphorylation or S1181D/E to mimic constitutive phosphorylation)

  • Include L1CAM disease-causing mutants (H210Q, I219T) as additional controls

3. Stimulation Paradigms:

  • Treat cells with NGF at different doses (10-100 ng/ml) and timepoints

  • Use recombinant L1CAM chimera proteins alongside NGF treatments

  • Consider dose-dependent experiments to assess robustness of the modulation

4. Assessment Methods:

  • Western blot analysis for TrkA phosphorylation status using phospho-specific antibodies

  • Immunoprecipitation to detect L1CAM-TrkA complexes

  • Neurite outgrowth assays to measure functional outcomes

  • Live-cell imaging to track receptor dynamics

5. Data Analysis:

  • Quantify the correlation between L1CAM S1181 phosphorylation levels and TrkA activation

  • Assess dose-dependency of the modulatory effect

  • Compare wild-type versus phosphomutant effects on TrkA signaling

This comprehensive approach will help elucidate whether S1181 phosphorylation is critical for L1CAM's ability to modulate TrkA receptor function and subsequent neurite outgrowth.

What are the best quantification methods for analyzing Phospho-L1CAM (S1181) in neuronal populations with varying expression levels?

Quantifying Phospho-L1CAM (S1181) in heterogeneous neuronal populations requires sensitive and versatile methods that can account for varying expression levels. Based on research practices in the field, the following methodological approaches are recommended:

For Tissue Section Analysis:

  • Standardized Sampling: Use non-serial sections (e.g., at least two sections spaced >100 μm apart) to avoid counting the same neurons twice

  • Cell Counting Parameters: Count neurons with visible nuclei to ensure consistent identification

  • Population Normalization: Express positive neurons as a percentage of the total neuronal population (minimum 400 neurons counted per section)

For Intensity-Based Quantification:

  • Confocal Microscopy Settings: Use consistent acquisition parameters including laser power, gain, and pinhole settings

  • Image Processing Protocol:

    • Apply standardized thresholding treatments

    • Quantify both area and number of immunoreactive profiles

    • Use software like ImageJ for consistent analysis

  • Internal Controls: Include reference regions or cell types with known expression levels

For Western Blot Quantification:

  • Loading Controls: Use appropriate loading controls (β-actin, GAPDH) and normalize phospho-signal

  • Phosphorylation-Specific Controls:

    • Include phosphatase-treated samples as negative controls

    • Use phosphopeptide competition assays to confirm specificity

  • Densitometry Analysis: Use linear range of detection and avoid saturated signals

Advanced Approaches for Heterogeneous Populations:

  • Single-Cell Analysis: Consider flow cytometry or single-cell Western approaches for heterogeneous populations

  • Multiplex Immunofluorescence: Combine with neuronal subtype markers to differentiate expression in distinct neuronal populations

  • Spatial Analysis: Implement spatial distribution analysis to identify region-specific variations

The combination of these approaches allows for robust quantification of Phospho-L1CAM (S1181) across neuronal populations with varying expression levels while maintaining scientific rigor.

How does phosphorylation at S1181 regulate L1CAM's role in axonal growth and neuronal development?

Phosphorylation at S1181 represents a critical regulatory mechanism for L1CAM's function in axonal growth and neuronal development. Research evidence indicates that this modification influences L1CAM's molecular interactions, membrane dynamics, and signaling capabilities.

The motility on the plasma membrane and binding ability of L1CAM are crucial modulatory mechanisms for axon growth, and these behaviors in cell-surface L1CAM are regulated by phosphorylation of the cytosolic domain . Specifically, S1181 phosphorylation appears to influence:

  • Conformational Changes: Phosphorylation at S1181 induces conformational alterations in L1CAM's cytoplasmic domain, changing its interaction capabilities with cytoskeletal components and signaling molecules

  • Sequential Phosphorylation Cascade: S1181 phosphorylation is required for subsequent phosphorylation of T1172 by CKII, establishing a regulatory cascade that controls L1CAM function

  • Growth Cone Dynamics: The phosphorylation state affects L1CAM's interaction with the cytoskeleton, influencing growth cone motility and directionality during axonal extension

  • Signaling Integration: Phosphorylated L1CAM at S1181 interfaces with other signaling pathways, particularly the NGF-TrkA pathway, which is crucial for neuronal development and differentiation

In experimental models, manipulation of S1181 phosphorylation through mutation (S1181A) affects neurite outgrowth capacity, suggesting direct functional consequences of this modification. Understanding this phosphorylation event provides insights into the molecular mechanisms governing neuronal development and may offer therapeutic targets for neurodevelopmental disorders associated with L1CAM dysfunction.

What is the relationship between L1CAM S1181 phosphorylation and neuropathic pain mechanisms?

Recent research has uncovered important connections between L1CAM S1181 phosphorylation and neuropathic pain mechanisms, particularly through axo-axonic synaptic reorganization processes. The evidence suggests several key relationships:

  • Morphologic Changes in Injured C-fiber Terminals: Phosphorylated L1CAM is associated with morphologic changes in injured C-fiber terminals, leading to the formation of hypertrophic varicosities that contribute to pain hypersensitivity

  • Synaptic Reorganization: Following nerve injury, phosphorylated L1CAM mediates aberrant axo-axonic synaptic reorganization, a process that underlies persistent neuropathic pain conditions

  • Quantifiable Changes in Dorsal Horn: After spared nerve injury (SNI), significant changes occur in the size and number of L1CAM-immunoreactive profiles in the dorsal horn, correlating with pain behavior development

  • Interaction with α2δ-1: The induction of α2δ-1 (a calcium channel subunit and therapeutic target for neuropathic pain) appears to interact with phosphorylated L1CAM pathways following nerve injury, suggesting convergent mechanisms

Methodologically, these relationships can be studied through:

  • Confocal microscopy of L1CAM-immunoreactive profiles in the dorsal horn

  • Quantification of area and number of immunoreactive profiles using image thresholding

  • Correlation of L1CAM phosphorylation with behavioral measurements of pain sensitivity

This emerging understanding of L1CAM's role in neuropathic pain offers potential new therapeutic targets for intervention, particularly those that might modulate L1CAM phosphorylation states.

How is Phospho-L1CAM (S1181) antibody utilized in cancer research, particularly for small cell lung cancer (SCLC)?

Phospho-L1CAM (S1181) antibodies have emerging applications in cancer research, with recent studies highlighting their particular relevance to small cell lung cancer (SCLC). L1CAM is expressed at higher levels in SCLC cell lines and tissues compared to lung adenocarcinoma and slightly higher than in adjacent normal tissues .

Research Applications in SCLC:

  • Subtyping and Classification:

    • L1CAM expression is notably high in the SCLC-N subtype

    • Phospho-L1CAM antibodies can help classify tumors based on L1CAM phosphorylation status

  • Therapeutic Target Development:

    • Anti-L1CAM monoclonal antibodies (e.g., HSL175) have been developed for antibody-drug conjugates (ADCs)

    • Phospho-specific antibodies help assess the phosphorylation state of L1CAM in tumor cells, which may affect internalization and therapeutic efficacy

  • Mechanism Studies:

    • Investigation of L1CAM's role in tumor progression, particularly in the context of phosphorylation-dependent functions

    • Understanding phosphorylation-specific effects on tumor cell migration, invasion, and metastasis

Methodological Approaches:

  • RNA Interference Assays:

    • Transient knockdown of L1CAM using siRNA (e.g., Silencer Select Pre-Designed siRNAs)

    • Verification of knockdown by RT-PCR and Western blot

  • Antibody Internalization Studies:

    • Assessment of antibody internalization with conjugates like DT3C

    • Evaluation of phosphorylation-dependent internalization mechanisms

  • Therapeutic Response Assessment:

    • Measuring apoptotic markers like cleaved PARP after treatment with anti-L1CAM therapeutics

    • Correlating phosphorylation status with response to L1CAM-targeted therapies

These applications demonstrate the significance of Phospho-L1CAM (S1181) antibodies in advancing our understanding of SCLC biology and developing targeted therapeutic approaches.

How might the interplay between L1CAM S1181 phosphorylation and cardiotoxicity mechanisms inform cancer treatment strategies?

Recent discoveries about L1CAM's role in cardiotoxicity present intriguing implications for cancer treatment strategies. L1CAM appears to be at the intersection of DNA damage responses, vascular integrity, and cardiac function, particularly in the context of anticancer therapies like thoracic irradiation and doxorubicin (Dox) treatment.

Research indicates that vascular endothelial cells with persistent DNA damage induced by irradiation and Dox treatment exhibit a fibrotic phenotype (endothelial-mesenchymal transition, EndMT) correlating with the colocalization of L1CAM and persistent DNA damage foci . This suggests a mechanism whereby L1CAM phosphorylation status may mediate cardiotoxicity.

Key Research Findings:

  • Anti-L1CAM antibody (Ab417) decreases L1CAM overexpression and nuclear translocation following DNA damage

  • Ab417 prevents cardiac dysfunction and prolongs survival after whole-heart irradiation or Dox treatment

  • Ab417 promotes endocytic internalization and lysosomal degradation of L1CAM

  • Ab417 enhances anti-tumor effects in combination with radiation therapy and Dox treatment

Methodological Implications for Cancer Treatment:

  • Dual-Target Approach: Targeting both tumor cells and protecting cardiac tissue through L1CAM modulation

  • Biomarker Development: Using phospho-L1CAM as a potential biomarker for cardiotoxicity risk

  • Combination Therapy Design: Incorporating anti-L1CAM antibodies with conventional therapies to reduce cardiotoxicity while enhancing anti-tumor effects

This research direction suggests that understanding the phosphorylation-dependent functions of L1CAM could lead to therapies that simultaneously protect cardiac tissue and enhance cancer treatment efficacy. Future studies should investigate whether S1181 phosphorylation specifically is involved in these cardioprotective mechanisms, and whether phospho-specific antibodies might have therapeutic potential.

What are the methodological considerations for studying the sequential phosphorylation cascade involving S1181 and T1172 in L1CAM?

Investigating the sequential phosphorylation cascade involving S1181 and T1172 in L1CAM requires sophisticated methodological approaches that can detect and manipulate phosphorylation events with high temporal and spatial resolution. Research has demonstrated that S1181 phosphorylation is required for subsequent T1172 phosphorylation by casein kinase II (CKII) , establishing a complex regulatory mechanism.

Advanced Methodological Approaches:

  • Site-Directed Mutagenesis Strategy:

    • Generate single mutants (S1181A, T1172A) and double mutants to dissect the sequential requirements

    • Create phosphomimetic mutants (S1181D/E, T1172D/E) to simulate constitutive phosphorylation

    • Compare wildtype with mutants in functional assays to assess biological consequences

  • Temporal Phosphorylation Analysis:

    • Employ pulse-chase phosphorylation assays with radioactive ATP or phosphate

    • Use kinase inhibitors with temporal administration to block specific steps in the cascade

    • Utilize time-resolved mass spectrometry to track phosphorylation kinetics

  • Structural Biology Approaches:

    • Implement hydrogen-deuterium exchange mass spectrometry (HDX-MS) to assess conformational changes induced by phosphorylation

    • Use nuclear magnetic resonance (NMR) to study structural dynamics of phosphorylated versus non-phosphorylated L1CAM cytoplasmic domains

    • Employ small-angle X-ray scattering (SAXS) for solution structure determination of different phospho-states

  • Advanced Microscopy Techniques:

    • Apply Förster resonance energy transfer (FRET) biosensors to monitor phosphorylation events in real-time

    • Implement super-resolution microscopy to visualize spatial distribution of phosphorylated forms

    • Use correlative light and electron microscopy (CLEM) to connect phosphorylation events with ultrastructural features

  • Validation Controls:

    • Employ epitope mapping with phospho-specific antibodies to confirm accessibility changes

    • Use phosphatase treatments as negative controls

    • Include peptide competition assays to verify antibody specificity to different phospho-states

By combining these methodologies, researchers can systematically unravel the complex interplay between S1181 and T1172 phosphorylation and their functional consequences for L1CAM activity.

What emerging technologies might enhance detection sensitivity and specificity for phosphorylated L1CAM in complex neural tissues?

Detecting phosphorylated L1CAM in complex neural tissues presents significant challenges due to cellular heterogeneity, low abundance of phosphorylated species, and complex three-dimensional architecture. Several emerging technologies offer promising advances to overcome these limitations:

1. Advanced Mass Spectrometry Approaches:

  • Targeted Proteomics: Multiple reaction monitoring (MRM) or parallel reaction monitoring (PRM) for sensitive detection of specific phosphopeptides

  • Single-Cell Phosphoproteomics: Emerging techniques for analyzing phosphorylation states at the single-cell level

  • Imaging Mass Spectrometry: MALDI-MSI for spatial mapping of phosphorylated proteins within tissue sections

2. Enhanced Immunological Methods:

  • Proximity Ligation Assays (PLA): Detection of phosphorylated L1CAM with 100-fold increased sensitivity over conventional immunohistochemistry

  • Quantum Dot-Based Immunofluorescence: Photostable nanoparticle-antibody conjugates with improved signal-to-noise ratios

  • Highly-Multiplexed Immunofluorescence: Cyclic immunofluorescence or mass cytometry imaging for simultaneous detection of multiple markers

3. Genetically-Encoded Biosensors:

  • FRET-Based Phosphorylation Sensors: Genetically encoded sensors that report on phosphorylation status in real-time

  • Split Fluorescent Protein Systems: Designed to reassemble upon specific phosphorylation events

  • Optogenetic Reporters: Light-sensitive domains coupled to phosphorylation-dependent conformational changes

4. Advanced Microscopy Technologies:

  • Expansion Microscopy: Physical enlargement of specimens to improve spatial resolution of phosphorylation sites

  • Light-Sheet Microscopy: Rapid imaging of large tissue volumes with reduced photobleaching

  • Super-Resolution Phosphoproteomics: Combining super-resolution microscopy with phospho-specific antibodies

5. Computational Methods for Enhanced Analysis:

  • Machine Learning Algorithms: For automated detection and quantification of phosphorylated L1CAM in large datasets

  • Spatial Transcriptomics Integration: Combining phosphoproteomic data with spatial transcriptomic information

  • Digital Pathology Tools: Automated image analysis platforms for consistent quantification across studies

These emerging technologies promise to revolutionize our ability to detect and characterize phosphorylated L1CAM in complex neural tissues, enabling more precise understanding of its spatiotemporal regulation and function in both normal neurobiology and pathological conditions.

What are the optimal storage and handling conditions for maintaining Phospho-L1CAM (S1181) antibody activity and specificity?

Maintaining the activity and specificity of Phospho-L1CAM (S1181) antibodies requires careful attention to storage and handling conditions. Based on manufacturer recommendations and research practices, the following guidelines should be followed:

Storage Conditions:

  • Long-term Storage: Store at -20°C or -80°C for maximum stability and shelf-life

  • Working Stock: For frequent use, small aliquots can be stored at 4°C for up to one month

  • Buffer Composition: Antibodies are typically supplied in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide, which helps maintain stability

Handling Best Practices:

  • Avoid Freeze-Thaw Cycles: Repeated freezing and thawing significantly reduces antibody activity; prepare single-use aliquots upon receipt

  • Temperature Transitions: Allow antibodies to equilibrate to room temperature before opening to prevent condensation

  • Sterile Technique: Use sterile pipette tips and tubes when handling antibodies to prevent contamination

  • Centrifugation: Briefly centrifuge antibody vials before opening to collect liquid at the bottom of the tube

Working Dilution Preparation:

  • Diluent Selection: Use high-quality, filtered buffers with appropriate blocking agents (e.g., BSA, non-fat dry milk)

  • Dilution Series: When optimizing, prepare a range of dilutions around the manufacturer's recommended concentration (e.g., 1:250, 1:500, 1:1000, 1:2000 for Western blot)

  • Storage of Diluted Antibody: Store diluted working solutions at 4°C and use within 24-48 hours for maximum sensitivity

Quality Control Procedures:

  • Positive Controls: Include known positive samples in each experiment to verify antibody activity

  • Specificity Controls: Use phosphopeptide competition or phosphatase-treated samples to confirm phospho-specificity

  • Batch Testing: When receiving a new lot, compare performance with previous lots using standardized samples

Adherence to these storage and handling guidelines will help ensure consistent experimental results and maximize the useful life of Phospho-L1CAM (S1181) antibodies.

How can researchers effectively troubleshoot non-specific binding or weak signals when using Phospho-L1CAM (S1181) antibodies?

When working with Phospho-L1CAM (S1181) antibodies, researchers may encounter challenges with non-specific binding or weak signals. Here are systematic troubleshooting approaches to address these common issues:

For Non-Specific Binding:

  • Optimize Blocking Conditions:

    • Increase blocking reagent concentration (3-5% BSA or milk)

    • Extend blocking time (1-2 hours at room temperature or overnight at 4°C)

    • Consider alternative blocking agents (casein, fish gelatin, commercial blocking buffers)

  • Adjust Antibody Conditions:

    • Increase antibody dilution (e.g., from 1:500 to 1:1000 for Western blot)

    • Reduce incubation temperature (4°C instead of room temperature)

    • Add 0.1-0.3% Tween-20 to antibody dilution buffer

  • Implement Validation Controls:

    • Perform peptide competition assays with phosphopeptides

    • Include phosphatase-treated samples as negative controls

    • Use knockout/knockdown samples to confirm specificity

  • Membrane/Slide Treatment:

    • For Western blots, increase washing steps duration and number

    • For IHC, optimize antigen retrieval methods (try Tris-EDTA pH 9.0)

    • Consider using specialized low-background membranes or slides

For Weak Signals:

  • Sample Preparation Optimization:

    • Ensure phosphatase inhibitors are included in lysis buffers

    • Verify protein concentration and loading amounts

    • Consider enrichment approaches (phosphoprotein enrichment columns)

  • Antibody Concentration Adjustment:

    • Decrease antibody dilution (e.g., from 1:2000 to 1:1000 for Western blot)

    • Extend primary antibody incubation time (overnight at 4°C)

    • Use signal enhancement systems (biotin-streptavidin, tyramide)

  • Detection System Enhancement:

    • Switch to more sensitive detection substrates (enhanced chemiluminescence)

    • Use fluorescent secondary antibodies with longer exposure times

    • Consider enzymatic amplification methods

  • Technical Considerations:

    • For Western blots, reduce transfer time or voltage to prevent protein over-transfer

    • For IHC, optimize fixation time and conditions

    • Ensure antibody is properly stored to maintain activity (see FAQ 5.1)

Decision Matrix for Common Problems:

ProblemPrimary CausesFirst InterventionsSecondary Approaches
High backgroundInsufficient blocking, antibody concentration too highIncrease blocking, increase antibody dilutionTry different blocking agent, reduce incubation temperature
No signalProtein not present, antibody inactive, detection failureConfirm protein expression, check positive control, verify detection systemTry antibody concentration series, enhance detection sensitivity
Multiple bandsCross-reactivity, protein degradation, post-translational modificationsIncrease antibody dilution, add protease inhibitors, verify phosphopeptide competitionUse gradient gels, optimize sample preparation, consider alternative antibody

By systematically applying these troubleshooting strategies, researchers can optimize their protocols for specific experimental conditions and achieve reliable, specific detection of phosphorylated L1CAM.

What considerations should guide the selection between polyclonal and monoclonal Phospho-L1CAM (S1181) antibodies for different research applications?

Selecting between polyclonal and monoclonal Phospho-L1CAM (S1181) antibodies requires careful consideration of the specific research application, experimental requirements, and technical trade-offs. Currently, most commercially available Phospho-L1CAM (S1181) antibodies are polyclonal , but understanding the differences is crucial for experimental design:

Comparative Analysis for Research Applications:

FeaturePolyclonal Phospho-L1CAM (S1181) AntibodiesMonoclonal Phospho-L1CAM (S1181) Antibodies
Epitope RecognitionRecognize multiple epitopes around S1181Target single epitope with higher specificity
SensitivityGenerally higher sensitivity due to multiple binding sitesMay have lower sensitivity but higher specificity
Batch-to-Batch VariationHigher variation between production lotsConsistent performance between batches
ProductionFaster production timelineLonger development process
CostGenerally lower costTypically more expensive
ApplicationsExcellent for IHC, WB, ELISAOptimal for standardized assays, therapeutic applications

Application-Specific Selection Guidance:

  • For Western Blot Analysis:

    • Polyclonals provide better sensitivity for detecting low-abundance phosphorylated L1CAM

    • Consider phosphopeptide competition controls to confirm specificity

    • Monoclonals may be preferred when absolute specificity is crucial

  • For Immunohistochemistry:

    • Polyclonals work well for fixed tissues with potentially altered epitopes

    • Optimize antigen retrieval methods (Tris-EDTA, pH9.0)

    • Monoclonals may reduce background in tissues with high endogenous immunoglobulins

  • For Functional Studies:

    • Consider whether epitope recognition might interfere with protein function

    • Monoclonals with defined epitopes may be preferable for functional blocking experiments

    • Recent developments like HSL175 (monoclonal anti-L1CAM) demonstrate potential for therapeutic applications

  • For Quantitative Assays:

    • Monoclonals provide more consistent results in standardized assays

    • Polyclonals may detect multiple phosphorylation states around S1181

    • Consider using both types to validate findings from complementary perspectives

  • For Novel Research Areas:

    • Polyclonals offer broader detection capability for exploratory research

    • Once specific epitopes of interest are identified, transition to monoclonals

    • Custom monoclonal development may be warranted for specialized applications

When selecting antibodies, researchers should always validate the chosen antibody with appropriate controls for their specific experimental system, regardless of whether they select a polyclonal or monoclonal variant.

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.