EML1 binds microtubules and regulates cytoskeletal organization during cortical development . Antibodies targeting EML1 are typically used in immunoblotting, immunofluorescence, and flow cytometry to study its expression, localization, and interactions. For example:
Immunofluorescence: Anti-EML1 antibodies (e.g., rabbit-derived) are used to visualize EML1 in progenitor cells of cerebral organoids .
Western blot: Antibodies like acetylated α-tubulin (AC-TUB) or β-actin are employed as loading controls in EML1 knockdown/knockout (KO) experiments .
While direct EML1 antibodies are not detailed in the sources, the following antibodies are critical in studying EML1-related pathways:
EML1 mutations are linked to neurodevelopmental disorders such as Usher syndrome type 1A . Antibodies play a role in:
Cerebral organoid models: EML1-KO organoids display ectopic neural rosettes and disrupted cytoskeletal structures, analyzed using ARL13B and PAX6 antibodies .
Primary cilia defects: ARL13B and acetylated tubulin antibodies reveal reduced cilia length in EML1-deficient cells .
Current research on EML1 antibodies is limited by:
Lack of specificity: Some studies rely on indirect markers (e.g., YAP1 for Hippo signaling) .
Technical challenges: Homozygous EML1-KO clones are prone to cell death, complicating antibody validation .
Common techniques involving antibodies in EML1 studies include:
EML1 (Echinoderm Microtubule Associated Protein Like 1) is a protein that modulates the assembly and organization of the microtubule cytoskeleton, playing a crucial role in regulating mitotic spindle orientation and the orientation of the plane of cell division . It is required for normal proliferation of neuronal progenitor cells in the developing brain and for normal brain development . Research has also demonstrated EML1's importance in retinal photoreceptor migration and survival , and its essential role in oocyte meiotic maturation . EML1 mutations have been associated with subcortical heterotopia in the brain, hydrocephalus, and cognitive impairment , making it a significant target for neurodevelopmental research.
EML1 antibodies are available in various formats that suit different research applications:
| Antibody Type | Host Species | Clonality | Common Applications | Typical Reactivity |
|---|---|---|---|---|
| Anti-EML1 polyclonal | Rabbit | Polyclonal | WB, IF, ICC, IHC(p) | Human, Mouse, Rat, Monkey |
| Anti-EML1 monoclonal | Mouse | Monoclonal (e.g., 5G3) | WB, ELISA | Human |
| Anti-EML1 C-terminal | Rabbit | Polyclonal | WB, ICC/IF | Human |
These antibodies target different epitopes of EML1, with some specifically recognizing the C-terminal region (aa 550 to C-terminus) or central regions (aa 100-200) of the protein .
According to published literature and antibody validation data, EML1 antibodies have been successfully used in multiple applications:
Western Blot (WB): Most validated application, with dilution ranges typically from 1:500 to 1:6000
Immunofluorescence (IF)/Immunocytochemistry (ICC): Successfully used to visualize EML1 localization in cells, particularly on spindle structures during cell division
Immunohistochemistry (IHC): Validated for tissue sections, especially for brain and retinal tissues
ELISA: Used in some antibody formulations, primarily for protein quantification
The specific application determines which antibody format is most appropriate, with polyclonal antibodies generally offering broader reactivity and monoclonal antibodies providing higher specificity .
For optimal Western blot detection of EML1:
Sample preparation: Use appropriate lysis buffers containing protease inhibitors to prevent degradation of EML1 (molecular weight ~90 kDa)
Gel selection: Use 7.5% SDS-PAGE gels which provide better resolution for the EML1 protein (observed MW: 90-92 kDa)
Transfer conditions: Optimize for larger proteins using lower voltage for longer time or semi-dry transfer systems
Antibody dilution: Start with manufacturer's recommended dilution (typically 1:1000-1:6000 for WB) and optimize based on signal-to-noise ratio
Detection system: Use enhanced chemiluminescence systems for sensitive detection
Controls: Include positive controls such as brain tissue lysates from mouse or rat, which show high expression of EML1
Loading control: Use β-actin as a validated loading control as shown in published EML1 research
Note that some commercial antibodies may only recognize specific isoforms of EML1. For example, one study reported that a polyclonal antibody (PA5-30016) recognized only the short EML1 isoform (~85-89 kDa) but not the long isoform .
For successful immunofluorescence staining of EML1:
Fixation method: Use paraformaldehyde (PFA) fixation (typically 4%) with Triton X-100 permeabilization for cultured cells
Blocking: Block with 3-5% BSA or normal serum (from the species of secondary antibody) for 1 hour at room temperature
Primary antibody: Apply EML1 antibody at appropriate dilution (typically 1:10-1:100 for IF/ICC) and incubate overnight at 4°C
Secondary antibody: Use fluorescently labeled secondary antibodies specific to the host species of the primary antibody
Nuclear counterstaining: Include DAPI or other nuclear stains to visualize nuclei
Mounting: Use anti-fade mounting medium to preserve fluorescence
Controls: Include both positive controls (tissues/cells known to express EML1) and negative controls (primary antibody omission)
Colocalization studies: Consider double-staining with microtubule markers (e.g., α-tubulin) to visualize EML1's association with microtubule networks
For specific applications like visualizing EML1 on the meiotic spindle in oocytes, specialized protocols have been published that demonstrate colocalization with other proteins like NUDC .
Validating antibody specificity is crucial for reliable results. For EML1 antibodies, consider these approaches:
Knockdown/knockout validation:
Use siRNA, shRNA, or morpholino oligos to reduce EML1 expression (as demonstrated in oocyte studies)
Compare antibody signal in control vs. knockdown samples by Western blot and immunostaining
Effective knockdown should show proportional reduction in signal intensity (e.g., 57.1% reduction was observed in morpholino studies)
Overexpression validation:
Peptide competition assay:
Pre-incubate the antibody with the immunizing peptide before application
Signal should be significantly reduced or eliminated
Multiple antibody validation:
Use different antibodies targeting different epitopes of EML1
Similar patterns support specificity
Expected subcellular localization:
EML1 demonstrates dynamic localization patterns during cell division that can be visualized using immunofluorescence techniques:
Cell cycle-dependent localization:
Experimental approach:
Synchronize cells at different cell cycle stages using cell cycle inhibitors
Fix cells and perform double immunostaining for EML1 and tubulin
Include cell cycle markers (e.g., phospho-histone H3 for mitosis)
Use confocal microscopy for high-resolution imaging
Drug perturbation experiments:
Live-cell imaging:
These approaches have revealed EML1's critical role in spindle organization and proper chromosome segregation.
To investigate EML1 protein interactions and complexes:
Co-immunoprecipitation (Co-IP):
Mass spectrometry-based approaches:
Proximity labeling methods:
Express EML1 fused to BioID or APEX2 to label proximal proteins in living cells
Identify interaction partners by streptavidin pulldown and mass spectrometry
Immunofluorescence co-localization:
Functional validation:
Gene enrichment analysis of EML1 interactors has revealed its involvement in multiple cellular processes including "RNA metabolism/splicing/localization," "cytoplasmic ribosomal subunit composition," and "protein folding and translation" .
EML1 has multiple isoforms, which presents challenges for antibody-based studies. To address this:
Isoform characterization:
Antibody selection:
Choose antibodies that recognize epitopes common to all isoforms for total EML1 detection
Select epitope-specific antibodies for studying particular isoforms
Be aware that some antibodies may preferentially detect certain isoforms (e.g., some commercial antibodies recognize only the short EML1 isoform)
Validation strategies:
Express recombinant isoforms and test antibody reactivity
Use tissues known to express specific isoforms as positive controls
Consider using tagged constructs of specific isoforms for overexpression studies
Genetic approaches:
Design isoform-specific knockdown/knockout strategies
Use precise gene editing techniques like CRISPR/Cas9 to target specific isoforms
When studying EML1 mutants, consider which isoforms are affected by the mutation
Interpreting contradictory results:
When results between studies differ, consider whether isoform specificity could explain the discrepancy
Compare the antibodies used and their known isoform recognition patterns
Some research has shown differences between in vivo mutant phenotypes and in vitro knockdown effects, possibly due to compensatory mechanisms involving other EML family members
Several models have been established for investigating EML1's role in neurodevelopment:
Mouse models:
The Eml1 tvrm360 mutant mouse exhibits subcortical heterotopia in the brain with associated hydrocephalus and cognitive impairment
This model is valuable for studying EML1's role in neuronal progenitor proliferation and migration
Limited availability of viable mutant mice may necessitate complementary approaches
In vitro neuronal cultures:
Primary neuronal cultures from wild-type and EML1 mutant animals
Neural progenitor cell cultures to study proliferation effects
Neurosphere assays to assess stem cell properties
Knockdown approaches:
Morpholino or siRNA-based knockdown in relevant cell types
In utero electroporation for spatiotemporally controlled knockdown in developing brain
Human cellular models:
iPSC-derived neural progenitors and organoids
CRISPR/Cas9-engineered cell lines with EML1 mutations
Patient-derived cells with naturally occurring EML1 mutations
Imaging techniques:
Immunohistochemistry with EML1 antibodies on brain sections
Live imaging of fluorescently labeled neural progenitors
Time-lapse microscopy to track migration and division orientation
When interpreting results, consider that phenotypic differences between models may arise from experimental context (acute vs. chronic loss) or compensatory mechanisms involving other EML family members .
EML1 antibodies have proven valuable in retinal research:
Expression pattern analysis:
Immunohistochemistry using anti-EML1 antibodies to map expression across retinal layers
Western blot analysis to quantify expression during development or in disease models
Functional studies:
Protein interaction studies:
Co-immunoprecipitation to identify retina-specific EML1 interaction partners
Co-localization studies with phototransduction proteins
Disease models:
Eml1 mutant mice show reduction in scotopic light response and mislocalized photoreceptors
Potential relevance to Usher syndrome type 1A, as EML1 has been identified as a candidate gene
Western blot analysis revealed reduced expression of phototransduction proteins (rhodopsin, transducin subunits, PDEγ) in Eml1 mutants
Technical considerations:
Research has shown that while EML1 is essential for photoreceptor migration and survival, it does not modulate phototransduction in mature rods and cones .
When EML1 antibodies are used as part of immunogenicity studies or when studying anti-drug antibodies:
Testing scheme design:
Data structure and analysis:
Controls and validation:
Include appropriate positive and negative controls
Perform assay validation including determination of cut-points
Assess potential drug interference
Interpretation challenges:
Statistical considerations:
Calculate incidence rates with appropriate denominators
Apply statistical methods for evaluating impact on efficacy and safety
Account for potential confounding factors
This approach enables proper evaluation of immunogenicity risk and definition of appropriate risk mitigation strategies .
Recent advances in computational antibody design offer promising approaches for developing novel EML1-targeting antibodies:
Epitope selection and analysis:
Identify accessible epitopes on EML1 structure using computational surface analysis
Target functionally important regions based on known biology
Consider epitope conservation across species if cross-reactivity is desired
Fragment-based design approach:
Computational optimization:
Optimize side-chain interactions between designed antibody and EML1 epitope
Enhance solubility and conformational stability through computational refinement
Predict binding affinity using molecular dynamics simulations
Experimental validation pipeline:
Advantages of this approach:
Fragment-based computational design has been successfully applied to generate antibodies against multiple targets, including the SARS-CoV-2 spike protein, and could be adapted for EML1-targeting applications .
Researchers may encounter several challenges when working with EML1 antibodies:
Additionally, when validating novel antibodies:
Compare staining patterns with published localization data (e.g., spindle localization during cell division)
Verify specificity through genetic approaches (knockdown/knockout)
For robust research outcomes, integrate EML1 antibody-based techniques with complementary approaches:
Multi-level protein analysis:
Combine Western blot (protein levels) with immunofluorescence (localization)
Supplement with mass spectrometry for unbiased protein identification and interactions
Use proximity labeling methods to identify proteins in the same cellular compartment
Functional validation:
Genetic approaches:
Live-cell imaging:
Complement fixed-cell antibody staining with live imaging of fluorescently tagged proteins
Track dynamic changes that might be missed in fixed samples
Validate that tagged protein behaves similarly to endogenous protein
Multi-omics integration:
Quantitative analysis:
Apply image analysis software for quantification of immunofluorescence signals
Use quantitative Western blot techniques with proper controls
Implement statistical analysis for rigorous interpretation