LAS1L antibodies are immunoglobulin proteins designed to specifically bind the LAS1L (LAS1-like ribosome biogenesis factor) protein, a nucleolar component required for 60S ribosomal subunit maturation and 28S rRNA processing . These antibodies are widely used in Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), and immunoprecipitation (IP) to investigate LAS1L's role in ribosome assembly, nucleolar integrity, and cancer biology .
LAS1L antibodies have been instrumental in identifying LAS1L's interaction with the mammalian Rix1 complex (PELP1, TEX10, WDR18, NOL9, SENP3), which co-sediments with pre-60S ribosomal particles . Key findings include:
Depletion of LAS1L or its interactors disrupts ITS-2 rRNA processing, leading to 32S pre-rRNA accumulation .
LAS1L complex proteins localize to the nucleolus and require active RNA polymerase I transcription for retention .
LAS1L antibodies are used to study its oncogenic roles:
Triple-negative breast cancer (TNBC): LAS1L overexpression correlates with tumor growth and metastasis. β-catenin inhibition reduces LAS1L levels, impairing nucleolar function .
p53-dependent cell cycle arrest: LAS1L depletion stabilizes p53, inducing G1 arrest, as shown in HCT116 and MCF7 cells .
LAS1L antibodies exhibit consistent performance across platforms:
| Application | Observed MW | Validated Cell Lines | Key Citations |
|---|---|---|---|
| Western Blot | 70–83 kDa | HEK293, HeLa, HCT116, MCF7 | |
| Immunofluorescence | Nucleolar | U2OS, HepG2, HeLa | |
| Immunoprecipitation | Pre-60S complex | HEK293, HCT116 |
LAS1L antibodies are used to explore therapeutic targets:
LAS1L (LAS1-like ribosome biogenesis factor) is a nucleolar protein essential for cell proliferation and ribosome biogenesis. In humans, the canonical protein has 734 amino acid residues with a molecular mass of 83.1 kDa . LAS1L is primarily localized in the nucleus and cytoplasm, with critical functions in the nucleolus. It belongs to the LAS1 protein family and plays a crucial role in the biogenesis of the 60S ribosomal subunit . LAS1L's importance is highlighted by the fact that its depletion leads to inhibition of rRNA processing, impaired synthesis of mature 28S rRNA, and ultimately affects cell proliferation through disruption of ribosome formation .
Several types of LAS1L antibodies are available for research applications, including both monoclonal and polyclonal variants. The most common forms are unconjugated antibodies, though some suppliers offer conjugated versions for specialized applications . Both rabbit and mouse-derived antibodies are available, with options like rabbit monoclonal anti-LAS1L antibody [EPR8988(B)] from suppliers like Abcam, which has been cited in published research . The reactivity of these antibodies varies, with most targeting human LAS1L, while some also recognize mouse and rat orthologs .
LAS1L antibodies are primarily used in several key applications:
Western Blot (WB): The most widely used application for detecting LAS1L protein expression and measuring protein levels in various experimental conditions .
Enzyme-Linked Immunosorbent Assay (ELISA): Used for quantitative analysis of LAS1L expression .
Immunocytochemistry (ICC) and Immunofluorescence (IF): Used to visualize the subcellular localization of LAS1L, particularly its nucleolar localization .
Immunoprecipitation (IP): Used to study protein-protein interactions involving LAS1L .
These techniques have been instrumental in establishing LAS1L's role in ribosome biogenesis and understanding its interaction network.
When designing experiments to study LAS1L's role in ribosome biogenesis, consider using a multi-faceted approach:
RNA interference (RNAi): Design experiments using shRNA or siRNA to deplete LAS1L. Previous studies have used sequences like 5'-CCGGGCTGCTACTTTGTCCTGGATTCTCGAGAATCCAGGACAAAGTAGCAGCTTTTTTG-3' cloned into pLKO.1-puro lentiviral vectors for effective knockdown .
rRNA processing analysis: Monitor pre-rRNA processing after LAS1L depletion, with particular focus on the 32S pre-rRNA intermediate, which accumulates in LAS1L-depleted cells, indicating processing defects in the internal transcribed spacer-2 (ITS-2) .
Cell cycle analysis: Include BrdU labeling with propidium iodide staining for FACS analysis to detect the G1 cell cycle arrest that occurs upon LAS1L depletion .
p53 pathway analysis: Monitor p53 and p21 levels through Western blotting to confirm the nucleolar stress response induced by LAS1L depletion .
Nucleolar integrity assessment: Use immunofluorescence with antibodies against nucleolar markers like UBF, fibrillarin, and nucleophosmin alongside LAS1L antibodies to assess nucleolar disruption .
When conducting immunofluorescence studies with LAS1L antibodies, include the following controls:
Negative controls:
Primary antibody omission: Perform the immunofluorescence protocol without the primary LAS1L antibody to assess background from secondary antibodies.
RNAi-depleted samples: Include cells where LAS1L has been depleted through RNAi to confirm antibody specificity .
Isotype controls: Use an irrelevant antibody of the same isotype as your LAS1L antibody.
Positive controls:
Technical controls:
Fixed cell samples should be permeabilized with 0.1% Triton X-100 for 10 minutes at room temperature to ensure antibody access to nuclear antigens .
Block samples with 1% BSA for 30 minutes to reduce non-specific binding .
Use appropriate fluorophore-conjugated secondary antibodies (e.g., Alexa Fluor 488 goat anti-rabbit or Alexa Fluor 647 goat anti-mouse) .
For optimal detection of LAS1L in Western blot experiments, follow these methodological guidelines:
Sample preparation:
Extract total protein from your samples using a buffer containing protease inhibitors.
For nuclear proteins like LAS1L, consider using nuclear extraction protocols for enrichment.
Gel selection:
Transfer conditions:
Transfer proteins to PVDF or nitrocellulose membranes using either wet or semi-dry transfer systems.
Blocking and antibody incubation:
Block membranes with 5% non-fat dry milk or BSA in TBST for 1 hour at room temperature.
Incubate with primary LAS1L antibody at manufacturer-recommended dilutions (typically 1:1000) overnight at 4°C.
Wash thoroughly with TBST before incubating with appropriate HRP-conjugated secondary antibody.
Detection:
Controls:
To investigate LAS1L protein interactions and complexes, consider these advanced approaches:
Co-immunoprecipitation (Co-IP):
Proximity-based labeling:
Consider BioID or APEX2-based approaches to identify proteins in close proximity to LAS1L in living cells.
Sucrose gradient fractionation:
Use this technique to separate ribosomal subunits and pre-ribosomal particles, then detect LAS1L in different fractions to understand its association with specific ribosomal assembly intermediates.
Chromatin Immunoprecipitation (ChIP):
If investigating LAS1L's potential role in transcriptional regulation, perform ChIP experiments to identify genomic binding sites.
CRISPR-Cas9 gene editing:
Generate cell lines with tagged endogenous LAS1L (e.g., FLAG, HA, or GFP tags) for purification of native complexes.
Validation of interactions:
To effectively study LAS1L's role in rRNA processing, employ the following specialized methods:
Northern blot analysis:
Pulse-chase labeling:
Use metabolic labeling with 32P or similar radioisotopes to track the kinetics of rRNA processing in control versus LAS1L-depleted cells.
rRNA quantification by qRT-PCR:
Fluorescence in situ hybridization (FISH):
Use probes targeting pre-rRNA species to visualize their localization and abundance in control versus LAS1L-depleted cells.
Ribosome profiling:
Analyze translational effects of LAS1L depletion through ribosome profiling to understand the functional consequences of disrupted ribosome biogenesis.
Ribosomal subunit analysis:
Use sucrose gradient centrifugation to quantify 40S and 60S ribosomal subunits and 80S monosomes in control versus LAS1L-depleted cells.
To investigate links between LAS1L dysfunction and human disease, consider these research approaches:
Mutation analysis:
Patient-derived cells:
Establish cell lines from patients with suspected LAS1L-related disorders to study protein expression, localization, and function.
CRISPR-Cas9 disease modeling:
Generate cell lines or model organisms with specific patient-derived LAS1L mutations to study their functional consequences.
Structure-function studies:
Map disease-associated mutations onto the functional domains of LAS1L to understand how they disrupt protein function.
Transcriptome and proteome analysis:
Compare gene expression and protein profiles between normal and LAS1L-mutant or depleted cells to identify dysregulated pathways.
Cell-based assays:
Develop rescue experiments where wildtype LAS1L is re-introduced into patient cells to determine if cellular phenotypes can be reversed.
Non-specific bands in LAS1L Western blots can arise from several sources:
Isoform detection:
Post-translational modifications:
LAS1L may undergo phosphorylation, ubiquitination, or other modifications that alter its migration pattern.
Antibody specificity issues:
Sample preparation problems:
Protein degradation during extraction can produce fragments that appear as additional bands
Incomplete denaturation can lead to aggregates or multimers
Technical solutions:
Increase washing stringency (higher salt or detergent concentration)
Optimize antibody dilution
Consider using monoclonal antibodies for higher specificity
Pre-absorb antibodies with common cross-reactive proteins
When using RNAi to study LAS1L function, be aware of these potential pitfalls:
Incomplete knockdown:
LAS1L is essential for cell proliferation, and cells with complete knockdown may be selected against, resulting in mixed populations with variable depletion levels.
Solution: Use inducible shRNA systems or optimize transfection/transduction efficiency.
Off-target effects:
Secondary effects versus direct effects:
Timing considerations:
Acute versus chronic depletion may yield different phenotypes.
Solution: Perform time-course experiments to distinguish primary from secondary effects.
Technical validation:
Distinguishing direct from indirect effects of LAS1L in ribosome biogenesis requires careful experimental design:
Temporal analysis:
Perform time-course experiments after LAS1L depletion to identify which defects appear first (likely direct effects) versus later (potentially secondary).
Structure-function studies:
Generate mutant LAS1L constructs affecting different functional domains to dissect which activities are essential for which aspects of ribosome biogenesis.
Protein-RNA interaction studies:
Use RNA immunoprecipitation (RIP) or crosslinking immunoprecipitation (CLIP) to identify direct RNA targets of LAS1L.
Proximity labeling approaches:
Employ BioID or APEX2 fusions with LAS1L to identify proteins in close proximity during ribosome synthesis.
In vitro reconstitution:
Attempt to reconstitute specific steps of ribosome assembly with purified components including LAS1L to test direct biochemical roles.
Conditional depletion systems:
Use rapid protein degradation systems (e.g., auxin-inducible degron) to achieve acute LAS1L depletion and observe immediate consequences.
LAS1L mutations have been implicated in several human diseases:
Intellectual developmental disorders:
Wilson-Turner syndrome (WTS):
Severe infantile conditions:
Potential cancer connections:
Given LAS1L's role in ribosome biogenesis and the link between dysregulated ribosome synthesis and cancer, further investigation into potential cancer associations is warranted, though direct evidence is currently limited.
Research approaches:
Genetic screening of affected populations
Functional characterization of patient-derived mutations
Animal models of LAS1L-associated diseases
Emerging molecular techniques that could advance LAS1L research include:
Cryo-electron microscopy:
Determine the structure of LAS1L alone and in complex with associated proteins and RNA to understand molecular mechanisms of action.
Integrative structural biology:
Combine X-ray crystallography, NMR, mass spectrometry, and computational modeling to build comprehensive structural models of LAS1L complexes.
Single-molecule approaches:
Use techniques like single-molecule FRET to study conformational changes and dynamics of LAS1L during ribosome assembly.
Spatial transcriptomics:
Apply techniques to visualize and quantify rRNA processing intermediates in situ to understand the spatial organization of ribosome assembly.
Proteomics:
Apply quantitative proteomics to identify changes in the composition of pre-ribosomal particles upon LAS1L depletion or mutation.
CRISPR screening:
Perform genome-wide CRISPR screens in LAS1L-depleted versus control cells to identify genetic interactions and compensatory pathways.
While therapeutic approaches for LAS1L-associated disorders are still in early stages, several potential strategies could be considered:
Gene therapy approaches:
For loss-of-function mutations, delivery of functional LAS1L cDNA could potentially restore normal protein function.
RNA-based therapeutics:
Antisense oligonucleotides or small interfering RNAs could be designed to modulate expression of specific LAS1L isoforms or target downstream effectors.
Small molecule screening:
Identify compounds that could stabilize mutant LAS1L protein or enhance residual function.
Targeting downstream pathways:
Ribosome-targeted therapies:
Explore the potential of compounds that modulate ribosome biogenesis or function to compensate for LAS1L dysfunction.
Personalized medicine approaches:
Develop patient-derived cellular models to test therapeutic strategies tailored to specific LAS1L mutations.