Recombinant monoclonal antibodies are validated for:
*Predicted mouse reactivity for 700712 .
LC3-II Detection: R&D Systems’ MAB85581 identifies LC3-II at ~15 kDa in HeLa cells treated with chloroquine, with knockout validation confirming specificity .
Tissue Distribution: Boster’s M01524 detects LC3B in human brain, liver, and rodent heart/brain lysates, aligning with known high expression in muscle and testis .
Isoform Discrimination: Boster’s A01524 (polyclonal) and M01524 (monoclonal) avoid cross-reactivity with LC3A/C isoforms, critical for accurate autophagy studies .
Post-translational Modifications: Thermofisher’s 700712 targets processed LC3-II, distinguishing it from unprocessed LC3-I (~18 kDa) .
Molecular Weight Variability: Observed LC3-II bands range from 15–18 kDa across antibodies, influenced by gel conditions and post-translational lipidation .
Storage Stability: Recombinant antibodies (e.g., 700712) require -20°C storage, while mouse monoclonals (e.g., AO1713a) tolerate 2–8°C .
The MAP1LC3B recombinant monoclonal antibody is produced through a rigorous and well-defined process. It begins with in vitro cloning, where the genes encoding both the heavy and light chains of the MAP1LC3B antibody are inserted into expression vectors. These vectors are then introduced into host cells, enabling the recombinant antibody's expression within a cell culture environment. Following expression, the MAP1LC3B recombinant monoclonal antibody undergoes purification from the supernatant of transfected host cell lines using affinity chromatography. Notably, this antibody exhibits high binding affinity for the human MAP1LC3B protein. It is also highly versatile, making it suitable for various applications, including ELISA, Western blotting, and flow cytometry.
MAP1LC3B is a pivotal protein in the autophagy pathway, playing a key role in autophagosome formation, cargo recognition, and autophagosome-lysosome fusion. It also participates in cellular quality control mechanisms. MAP1LC3B facilitates the removal of damaged organelles, misfolded proteins, and protein aggregates, contributing to the maintenance of cellular health and preventing the accumulation of potentially harmful substances.
MAP1LC3B (Microtubule-associated protein 1 light chain 3 beta) is a critical protein involved in autophagosome formation during autophagy. It serves as one of the most widely used markers for monitoring autophagy in research settings. MAP1LC3B belongs to the LC3 family, which comprises three highly homologous members: MAP1LC3A (LC3A), MAP1LC3B (LC3B), and MAP1LC3C (LC3C) . In mammalian cells, MAP1LC3B functions as an autophagy receptor essential for autophagosome elongation, making it a critical component for studying autophagy dynamics .
The protein exists in two distinct forms: the cytoplasmic LC3-I (18 kDa) and the lipidated LC3-II (16 kDa), which is generated during autophagosome and autophagolysosome formation . This conversion from LC3-I to LC3-II makes MAP1LC3B particularly valuable for monitoring autophagy flux in various experimental systems. When visualized through immunofluorescence techniques, LC3-II appears as distinctive punctate structures that represent autophagic vesicles, providing a visual signature of autophagic activity .
MAP1LC3B antibodies are versatile tools employed across multiple experimental techniques:
The experimental approach should be selected based on research questions, with appropriate positive controls included to validate antibody performance . For immunofluorescence experiments, paraformaldehyde/methanol fixation is often recommended to preserve autophagic structures .
Distinguishing between LC3-I and LC3-II forms is crucial for accurately interpreting autophagy dynamics:
Western blotting is the primary technique for differentiating between these forms, where LC3-I appears at approximately 18 kDa and LC3-II at 16 kDa . This apparent paradox in migration (the lipidated form migrates faster despite increased molecular weight) occurs due to the hydrophobicity imparted by phosphatidylethanolamine conjugation. PVDF membranes are strongly recommended over nitrocellulose for immunoblot analysis, as they provide better retention of the lipidated LC3-II form .
For optimal separation, researchers should:
Use 15-16% polyacrylamide gels or specialized gradient gels
Employ casein/Tween 20-based blocking buffers to reduce background
Include appropriate positive controls (such as an enriched cell fraction containing both LC3-I and LC3-II)
Consider normalization to housekeeping proteins when quantifying band intensities
In immunofluorescence studies, LC3-II appears as distinct punctate structures that represent autophagosomes, while LC3-I shows diffuse cytoplasmic staining. The number of puncta can be quantified to assess autophagy levels .
Robust experimental design for MAP1LC3B detection requires multiple complementary approaches:
For immunoblotting experiments:
Include both autophagy inducers (starvation, rapamycin) and inhibitors (bafilomycin A1, chloroquine) to assess autophagy flux rather than static LC3-II levels
Employ multiple time points to capture the dynamic nature of autophagy
Use PVDF membranes specifically for LC3B detection, as the lipidated form can be lost on nitrocellulose
Validate results with knockdown/knockout controls to confirm antibody specificity
For immunofluorescence/immunohistochemistry:
Optimize fixation protocols (paraformaldehyde/methanol) to preserve autophagosomal structures
Implement quantitative analysis of LC3 puncta formation
Include co-localization studies with other autophagosomal markers
Consider super-resolution microscopy for detailed analysis of autophagosomal structures
Cross-validation between different techniques provides the most comprehensive assessment of autophagic processes. Researchers should also standardize experimental conditions, including cell confluency and passage number, which can significantly impact basal autophagy levels.
The combined evaluation of MAP1LC3B and SQSTM1/p62 provides deeper insights into autophagy status in cancer tissues:
Recent research on breast invasive ductal carcinoma (IDC) found that tumor tissues show higher protein levels of both MAP1LC3B and cytoplasmic SQSTM1 compared to adjacent normal tissues . Interestingly, high levels of MAP1LC3B were associated with better disease-specific survival and disease-free survival (DFS) in IDC patients . Furthermore, high co-expression of MAP1LC3B and SQSTM1 was significantly associated with better DFS in these patients .
These findings challenge simplistic interpretations of autophagy markers in cancer. While increased SQSTM1/p62 typically indicates autophagy inhibition (as it accumulates when autophagy is impaired), and high MAP1LC3B can signal either increased autophagosome formation or blocked autophagosome-lysosome fusion, their combined expression patterns provide more nuanced information about tumor biology.
Researchers analyzing these markers should:
Evaluate both markers simultaneously rather than in isolation
Consider tissue-specific contexts when interpreting expression patterns
Correlate expression with clinical parameters and patient outcomes
Differentiate between cytoplasmic and nuclear SQSTM1/p62 localization, as they may have distinct functional implications
Addressing variability in antibody performance requires rigorous validation and standardization:
Recombinant monoclonal antibodies offer several advantages over traditional antibodies, including:
Increased sensitivity and confirmed specificity
High repeatability and excellent batch-to-batch consistency
Researchers should implement the following practices:
Validate antibody specificity using knockout/knockdown controls
Test multiple antibody clones when establishing new protocols
Include appropriate positive controls with each experiment (such as enriched cell fractions containing both LC3-I and LC3-II)
Standardize sample preparation, including lysis buffers and protein extraction methods
Determine optimal antibody dilutions empirically for each experimental system
Consider recombinant antibody formats for improved reproducibility
For immunohistochemistry applications, antigen retrieval methods should be carefully optimized, as MAP1LC3B epitopes can be sensitive to fixation and embedding procedures. Cross-validation with multiple antibodies targeting different epitopes can provide additional confidence in the specificity of staining patterns.
MAP1LC3B shows conservation across multiple species, enabling comparative studies with important considerations:
The available antibodies demonstrate cross-reactivity with MAP1LC3B from multiple species including human, mouse, rat, canine, and hamster . In zebrafish, the orthologous gene map1lc3b (ZDB-GENE-030131-1145) encodes a protein with similar predicted functions, including microtubule binding, phosphatidylethanolamine binding, and roles in macroautophagy .
When designing cross-species experiments:
Verify epitope conservation through sequence alignment before selecting antibodies
Validate antibody reactivity in each species experimentally
Consider species-specific expression patterns (for example, MAP1LC3B in zebrafish is expressed in cardiovascular system, central nervous system, liver, neural tube, and pleuroperitoneal region)
Adjust sample preparation protocols for tissue-specific differences
Implement controls specific to each model organism
For zebrafish studies specifically, researchers should note that map1lc3b has had several previous nomenclatures including Lc3, wu:fb60g11, and zgc:56434 . When comparing across species, researchers should carefully account for potential functional differences despite sequence homology.
Western blotting for MAP1LC3B requires specific technical considerations:
The detection of both LC3-I (18 kDa) and LC3-II (16 kDa) forms demands careful optimization of electrophoresis and transfer conditions. PVDF membranes are strongly recommended over nitrocellulose for LC3B immunoblotting, as they provide superior retention of the lipidated LC3-II form .
Key optimization strategies include:
Using higher percentage gels (15-16%) for better separation of closely migrating bands
Implementing casein/Tween 20-based blocking and blot incubation buffers to reduce background
Employing gradient gels when analyzing samples with varying protein sizes
Careful sample preparation, as LC3-II is sensitive to freeze-thaw cycles and proteolytic degradation
Loading equal protein amounts (15-30 μg total protein) per lane
Including positive controls such as enriched cell fractions containing both LC3-I and LC3-II
Quantification should consider the ratio of LC3-II to LC3-I or the ratio of LC3-II to a loading control such as β-actin or GAPDH, rather than absolute band intensities. To assess autophagic flux properly, researchers should include conditions with and without lysosomal inhibitors.
MAP1LC3B exhibits diverse functions depending on cellular location and context:
While primarily recognized for its role in autophagosome formation, MAP1LC3B has been identified in various cellular compartments with distinct functions:
In autophagy: MAP1LC3B promotes autophagosome formation and cargo recognition through interaction with adaptor proteins like SQSTM1/p62
In primary ciliogenesis: MAP1LC3B promotes this process by removing OFD1 from centriolar satellites via the autophagic pathway
In cellular stress response: MAP1LC3B participates in the response to nitrogen starvation and other cellular stressors
In cancer biology: MAP1LC3B expression patterns correlate with disease outcomes in various cancer types, with context-dependent prognostic implications
The functional diversity of MAP1LC3B highlights the importance of considering cellular context when interpreting experimental results. Researchers should employ compartment-specific markers and co-localization studies to discern the specific roles of MAP1LC3B in their experimental systems.
Analysis of MAP1LC3B in clinical samples requires specific methodological considerations:
When working with tissue microarrays or patient samples:
Standardize fixation protocols to ensure consistent preservation of autophagy-related structures
Implement appropriate antigen retrieval methods, as MAP1LC3B epitopes can be sensitive to fixation
Include both tumor and adjacent normal tissue for comparative analysis
Consider co-staining with SQSTM1/p62 for more comprehensive evaluation of autophagy status
Stratify samples based on relevant clinical parameters (stage, grade, treatment history)
Employ quantitative image analysis for objective assessment of staining patterns and intensities
Recent research using tissue microarrays from 274 breast invasive ductal carcinoma patients demonstrated that tumor tissues show higher protein levels of MAP1LC3B compared to adjacent normal tissues . Furthermore, high levels of MAP1LC3B were associated with better survival outcomes, highlighting the prognostic value of this marker .
When interpreting results, researchers should consider that autophagy is highly dynamic and tissue samples represent a single time point in this process. Additionally, the relationship between MAP1LC3B expression and patient outcomes appears to be cancer-type specific, requiring careful consideration of the disease context.
Emerging imaging technologies offer new opportunities for MAP1LC3B research:
Super-resolution microscopy techniques like STORM, PALM, and STED can reveal ultrastructural details of autophagosomes below the diffraction limit, providing insights into the spatial organization of MAP1LC3B during autophagosome formation. These techniques can be combined with MAP1LC3B antibodies to visualize:
The transition from diffuse to punctate LC3B structures during autophagy induction
The dynamic recruitment of LC3B to forming autophagosomes
Co-localization with other autophagy proteins at nanoscale resolution
Structural changes in autophagosomes during maturation
Live-cell imaging approaches using fluorescently-tagged MAP1LC3B constructs complemented with validated antibodies for fixed-cell analysis can provide temporal information about autophagy dynamics. Advanced quantitative image analysis algorithms can extract multi-parametric data from MAP1LC3B staining patterns, including puncta size, intensity, distribution, and co-localization with other markers.
Future directions may include correlative light and electron microscopy (CLEM) to link fluorescently labeled MAP1LC3B to ultrastructural features of autophagosomes, and multiplexed imaging to simultaneously visualize multiple components of the autophagy machinery.
MAP1LC3B research extends to numerous disease contexts:
While much research has focused on MAP1LC3B in cancer, this protein plays critical roles in various pathological conditions:
Neurodegenerative diseases: Altered MAP1LC3B processing has been implicated in Alzheimer's, Parkinson's, and Huntington's diseases, where impaired autophagy contributes to protein aggregation
Cardiovascular disorders: MAP1LC3B-mediated autophagy influences cardiac remodeling and response to ischemia-reperfusion injury
Infectious diseases: MAP1LC3B participates in xenophagy, the autophagic clearance of intracellular pathogens
Metabolic disorders: Dysregulated MAP1LC3B processing affects lipid metabolism and cellular energy homeostasis
Future research should focus on tissue-specific and context-dependent roles of MAP1LC3B in these conditions. Understanding how MAP1LC3B function varies across tissues and disease states may reveal new therapeutic opportunities. The development of tissue-specific MAP1LC3B antibodies or those that selectively recognize specific post-translational modifications could advance understanding of MAP1LC3B's diverse roles.