LAMP2 (lysosomal-associated membrane protein 2), also known as CD107b, is a transmembrane glycoprotein critical for lysosomal function, autophagy, and immune responses. Recombinant monoclonal antibodies targeting LAMP2 are engineered via in vitro cloning and expression systems, offering high specificity and consistency compared to traditional hybridoma-derived antibodies . These antibodies are widely used in research and diagnostics to study lysosomal biology, autophagy pathways, and immune cell activation.
LAMP2 antibodies enable diverse experimental approaches:
Detection: Identifies LAMP2 in lysates (e.g., HepG2 cells) at ~120 kDa under reducing conditions .
Key Findings:
Localization: Detects lysosomal or plasma membrane LAMP2 in activated immune cells (e.g., cytotoxic T cells, mast cells) .
Clinical Relevance:
Intracellular Staining: Quantifies LAMP2 in human PBMCs or granular cells .
Limitations: High glycosylation may reduce antibody binding efficiency .
Chaperone-Mediated Autophagy (CMA): LAMP2 binds HSC70/HSP90 complexes, enabling substrate uptake into lysosomes .
Autophagosome Fusion: Required for STX17 recruitment and lysosome fusion .
MHCII Antigen Presentation: Facilitates exogenous antigen processing but not endogenous antigens .
Infection Pathways: Interacts with furin to process mumps virus fusion proteins .
The LAMP2 recombinant monoclonal antibody production is a meticulously controlled process. It begins with in vitro cloning, where the genes for both the heavy and light chains of the LAMP2 antibody are precisely incorporated into expression vectors. Subsequently, these modified vectors are introduced into host cells, providing an optimal environment for the expression of the recombinant antibody within a cell culture system. After expression, the LAMP2 recombinant monoclonal antibody undergoes a rigorous purification process using affinity chromatography. A key feature of this antibody is its specific binding affinity for the human LAMP2 protein. It is recommended for use in ELISA and IHC applications.
LAMP2 plays a crucial role in the proper functioning of lysosomes, essential for cellular waste disposal, recycling of cellular components, and various other cellular processes. Dysfunction or deficiency of LAMP2 can lead to lysosomal storage diseases and other health conditions.
LAMP2 (also known as CD107b) is a transmembrane glycoprotein that serves as a major component of lysosomal membranes. Mature human LAMP2 consists of a 347 amino acid intralumenal domain, a 24 amino acid transmembrane segment, and a 35 amino acid cytoplasmic tail . The protein plays critical roles in multiple cellular processes including:
Lysosomal biogenesis and pH regulation
Chaperone-mediated autophagy
Autophagosome-lysosome fusion
Protein degradation pathways
Antigen presentation in immune cells
LAMP2 functions as a direct inhibitor of the proton channel TMEM175, facilitating lysosomal acidification required for optimal hydrolase activity . Its importance extends beyond basic lysosomal function, as it also participates in immune cell activation and exosome biology . Mutations in LAMP2 cause Danon disease, characterized by cardiomyopathy, myopathy, and cognitive impairment, highlighting its physiological significance.
LAMP2 is an approximately 110 kDa transmembrane glycoprotein with several distinctive structural features:
The lumenal domain is organized into two heavily N-glycosylated regions separated by a Ser/Pro-rich linker carrying minor O-linked glycosylation
While the calculated molecular weight is approximately 45 kDa, the mature protein typically appears at 100-130 kDa on Western blots due to extensive glycosylation
The protein contains multiple disulfide bonds that maintain its tertiary structure
Alternative splicing generates multiple LAMP2 isoforms with substituted juxtamembrane lumenal regions, transmembrane segments, and cytoplasmic tails . The most well-characterized isoforms include:
LAMP2A: Critical for chaperone-mediated autophagy, functioning as a receptor for substrate proteins
LAMP2B: Predominantly expressed in cardiac and skeletal muscle
LAMP2C: Involved in RNA and DNA autophagy
Each isoform has unique C-terminal sequences that determine their specific functions and interactions within cellular pathways.
Several types of LAMP2 recombinant monoclonal antibodies are commercially available, differentiated by host species, clones, and target epitopes:
Many of these antibodies have been extensively validated and cited in numerous publications, with some clones like GL2A7 cited in over 260 scientific papers . When selecting an antibody, researchers should consider the target species compatibility, intended applications, and whether isoform specificity is required for their particular research question.
Selecting the appropriate LAMP2 antibody requires consideration of several key factors:
Target species compatibility: Confirm cross-reactivity with your species of interest. Most LAMP2 antibodies work well with human samples, but murine cross-reactivity varies by clone .
Application suitability: Different clones perform optimally in different applications. For example:
Epitope recognition: Consider whether the antibody recognizes:
All LAMP2 isoforms (most commercial antibodies)
Specific isoforms (required for specialized studies of LAMP2A, B, or C functions)
Conformational vs. linear epitopes (affects performance in denatured vs. native conditions)
Validation evidence: Review manufacturer data showing antibody specificity through:
The most reliable approach is to test multiple antibodies in preliminary experiments with appropriate positive and negative controls relevant to your experimental system.
Detecting LAMP2 by Western blotting requires specific considerations due to its heavily glycosylated nature and membrane localization:
Sample preparation:
Use RIPA or NP-40 buffer supplemented with protease inhibitors
Include 1-2% SDS for complete solubilization of membrane proteins
Add phosphatase inhibitors if phosphorylation status is important
Sonicate briefly to shear DNA and reduce sample viscosity
Protein loading and separation:
Transfer conditions:
Employ wet transfer rather than semi-dry for complete transfer of large proteins
Use PVDF membranes (0.45 μm pore size) for better protein retention
Transfer at lower voltage (30V) overnight at 4°C for improved efficiency
Blocking and antibody incubation:
Detection considerations:
This protocol has been validated with multiple LAMP2 antibodies including ARC54762, H4B4, and GL2A7 across various cell types .
For successful immunofluorescence detection of LAMP2:
Cell preparation and fixation:
Culture cells on glass coverslips or chamber slides to 70-80% confluence
Fix with 4% paraformaldehyde for 15-20 minutes at room temperature
For optimal lysosomal preservation, avoid methanol fixation which can distort vesicular structures
Permeabilization options:
For total LAMP2: Use 0.1-0.5% saponin (preferred for lysosomal proteins over Triton X-100)
For surface LAMP2 only: Omit permeabilization step
Blocking and antibody incubation:
Detection and counterstaining:
Use fluorophore-conjugated secondary antibodies at manufacturer-recommended dilutions
Include DAPI or Hoechst for nuclear counterstaining
Mount with anti-fade medium containing glycerol or similar agents
Imaging considerations:
Expect punctate cytoplasmic staining pattern consistent with lysosomal localization
Use confocal microscopy for optimal resolution of lysosomal structures
Consider co-staining with other lysosomal markers (e.g., LAMP1) for validation
For co-localization studies, LAMP2 antibodies from rat (GL2A7) can be combined with antibodies from different species targeting other proteins of interest . Z-stack acquisition is recommended for comprehensive analysis of lysosomal distribution throughout the cell volume.
Flow cytometric analysis of LAMP2 requires consideration of whether total or surface expression is being measured:
Cell preparation:
Harvest cells in exponential growth phase
Prepare single-cell suspensions (1-5 × 10^6 cells/ml)
Wash in PBS containing 2% FBS (flow buffer)
For total LAMP2 detection (most common):
Fix cells with 2-4% paraformaldehyde for 10-15 minutes
Permeabilize with 0.1% saponin in flow buffer (maintain saponin in all subsequent steps)
Block with 5% normal serum from secondary antibody species
For surface LAMP2 detection (activation marker):
Use unfixed cells or gentle fixation (1% paraformaldehyde, 5 minutes)
Omit permeabilization step
Keep cells cold (4°C) throughout the procedure
Antibody staining:
Controls and analysis:
Include isotype control matched to primary antibody concentration
Prepare single-color controls for compensation if performing multicolor analysis
Gate on live cells using viability dye if analyzing fixed/permeabilized samples
Analyze LAMP2 as median fluorescence intensity rather than percent positive
This protocol has been validated using various LAMP2 antibodies including clone 743320 and H4B4 . For detection of LAMP2 in HeLa cells, paraformaldehyde fixation followed by saponin permeabilization has shown robust results with minimal background .
Chaperone-mediated autophagy (CMA) is a selective form of autophagy where LAMP2A serves as the lysosomal receptor. LAMP2 antibodies can be instrumental in studying this process:
Isoform-specific detection:
If using general LAMP2 antibodies, complement with LAMP2A-specific antibodies when available
Monitor LAMP2A multimerization (the active form for CMA) via non-reducing gels
Track changes in LAMP2A levels during starvation or stress conditions
Co-immunoprecipitation studies:
Microscopy-based approaches:
Perform immunofluorescence to visualize LAMP2 and CMA substrates
Analyze colocalization of substrates with LAMP2-positive lysosomes
Conduct live-cell imaging with fluorescently tagged LAMP2 and substrate proteins
Functional CMA assays:
Use LAMP2 antibodies to monitor changes in CMA activity under various conditions
Combine with lysosomal isolation techniques to study direct protein translocation
Assess the impact of manipulating LAMP2 expression on degradation of known CMA substrates
LAMP2 functions by binding target proteins and targeting them for lysosomal degradation . In this process, it acts downstream of chaperones like HSPA8/HSC70, which recognize and bind substrate proteins and mediate their recruitment to lysosomes . Quantitative analysis of LAMP2-substrate interactions provides valuable insights into CMA regulation in various physiological and pathological contexts.
LAMP2 has been identified as an endocytic receptor on human dendritic cells that routes cargo into immunogenic exosomes. LAMP2 antibodies can be used to study this pathway:
Tracking receptor-mediated antigen uptake:
Conjugate antigens directly to anti-LAMP2 antibodies (e.g., clone H4B4)
Compare with other targeting strategies (e.g., DEC-205, DC-SIGN)
Monitor internalization kinetics and subcellular trafficking
Analysis of antigen routing:
Use immunofluorescence microscopy to track LAMP2-bound antigens
Determine colocalization with HLA-DR in MIIC compartments
Compare surface presentation versus exosomal loading
Exosome characterization:
Isolate exosomes from dendritic cells treated with LAMP2-targeted antigens
Quantify antigenic content in exosomes versus cell-surface presentation
Assess T cell stimulatory capacity of these exosomes
Research has shown that surface LAMP-2 is rapidly internalized upon ligation and traffics to the MIIC compartment, similar to known DC endocytic receptors . Surprisingly, despite this trafficking pattern, monocyte-derived dendritic cells pulsed with antigen conjugated to anti-LAMP-2 antibodies express fewer antigen-derived peptides in the HLA class II peptidome and evoke less T cell proliferation . Instead, antigens are selectively routed into highly immunogenic exosomes that stimulate robust CD4 T cell responses.
This pathway represents a novel mechanism for generating immunogenic extracellular vesicles that may contribute to immune responses both locally and at distant sites.
LAMP2 plays a critical role in autophagosome-lysosome fusion, and studying this function requires specific methodological approaches:
Colocalization analysis:
Use LAMP2 antibodies to mark lysosomes in fixed cells
Co-stain with autophagosome markers (LC3-II, ATG16L1)
Quantify colocalization under various conditions (starvation, drug treatments)
Employ super-resolution microscopy for detailed analysis of fusion sites
Molecular mechanism investigation:
Examine STX17 accumulation on autophagosomes in LAMP2-deficient cells
Analyze VAMP8 and other SNARE proteins in relation to LAMP2
Study the impact of LAMP2 mutations on fusion efficiency
Functional autophagy assays:
Compare autophagy flux in LAMP2-depleted versus control cells
Measure LC3-II accumulation with/without lysosomal inhibitors
Assess degradation of long-lived proteins or specific autophagy substrates
Live-cell imaging approaches:
Use fluorescently-tagged LAMP2 constructs with autophagosomal markers
Monitor fusion events in real-time
Calculate fusion rates under various experimental conditions
Research has revealed that cells lacking LAMP2 express normal levels of VAMP8 but fail to accumulate STX17 on autophagosomes, which is likely the explanation for the defective fusion between autophagosomes and lysosomes in these cells . This finding highlights LAMP2's role in recruiting or stabilizing key fusion machinery components, providing mechanistic insight into its function in autophagy.
Western blotting for LAMP2 can present several challenges due to its extensive glycosylation and membrane localization:
For optimal results, include validated positive control lysates such as HeLa, U937, or Jurkat cells, which are known to express detectable levels of LAMP2 . Consider non-reducing conditions for some antibodies, as certain epitopes may be conformation-dependent.
Proper controls are essential for generating reliable and interpretable results with LAMP2 antibodies:
Positive controls:
Negative controls:
Specificity controls:
Peptide competition assay (pre-incubation of antibody with immunizing peptide)
Validation with multiple antibodies targeting different LAMP2 epitopes
Deglycosylation controls to confirm identity of glycosylated bands
Application-specific controls:
For immunofluorescence: Co-staining with other lysosomal markers
For flow cytometry: Fluorescence minus one (FMO) controls
For Western blotting: Loading controls and molecular weight markers
Biological validation:
LAMP2 upregulation during starvation (autophagic response)
Surface LAMP2 increase upon activation of immune cells
Altered LAMP2 distribution after treatment with lysosomal inhibitors
Including these controls enables confident interpretation of results and troubleshooting of potential issues across different experimental platforms.
Distinguishing between total LAMP2 (predominantly lysosomal) and surface LAMP2 requires specific methodological approaches:
Flow cytometry approach:
Total LAMP2: Fix cells with paraformaldehyde and permeabilize with saponin or similar agent
Surface LAMP2: Stain unfixed cells at 4°C, or use very mild fixation without permeabilization
Compare median fluorescence intensity between permeabilized and non-permeabilized samples
Surface LAMP2 serves as an activation marker for certain cell types (T cells, mast cells, monocytes, platelets)
Microscopy-based distinction:
Total LAMP2: Standard fixation and permeabilization showing punctate cytoplasmic pattern
Surface LAMP2: Surface staining protocol showing membrane localization
Use confocal microscopy for accurate discrimination between surface and intracellular signals
Z-stack analysis to confirm genuine surface vs. intracellular localization
Biochemical separation:
Surface biotinylation followed by streptavidin pull-down to isolate surface proteins
Probe for LAMP2 in biotinylated (surface) and non-biotinylated (internal) fractions
Compare with total cell lysate to determine relative distribution
Kinetic analysis:
Monitor LAMP2 internalization from surface over time using antibody feeding assays
Track antibody-bound surface LAMP2 as it traffics to internal compartments
Quantify internalization rates under various experimental conditions
LAMP2 antibodies are increasingly employed in neurodegenerative disease research to investigate lysosomal and autophagic dysfunction:
Alzheimer's disease studies:
Quantifying LAMP2 expression in brain tissue from patients
Examining colocalization of amyloid-β and tau with LAMP2-positive lysosomes
Investigating impaired lysosomal clearance mechanisms
Parkinson's disease applications:
Studying α-synuclein processing in LAMP2-positive compartments
Analyzing chaperone-mediated autophagy defects in dopaminergic neurons
Investigating LAMP2A expression in relation to disease progression
Lysosomal storage disorders:
Danon disease (caused by LAMP2 mutations) research
Using LAMP2 as a marker for lysosomal accumulation in various storage disorders
Tracking therapeutic efficacy of treatments targeting lysosomal function
Therapeutic development:
Screening compounds that enhance LAMP2 expression or function
Monitoring autophagy modulation using LAMP2 as a readout
Developing strategies to upregulate specific LAMP2 isoforms
LAMP2 plays a vital role in chaperone-mediated autophagy, which is increasingly recognized as dysfunctional in various neurodegenerative conditions . Methodological approaches include immunohistochemistry of post-mortem brain tissue, primary neuronal cultures, patient-derived iPSCs, and animal models of neurodegeneration.
LAMP2 antibodies provide valuable tools for investigating exosome biology, particularly in immune contexts:
Exosome characterization:
Western blotting for LAMP2 as an exosomal marker
Immunogold labeling with LAMP2 antibodies for electron microscopy
Flow cytometric analysis of LAMP2-positive extracellular vesicles
Selective exosome isolation:
Immunoaffinity capture using anti-LAMP2 antibodies
Characterizing LAMP2-positive vs. LAMP2-negative exosome populations
Correlating LAMP2 content with exosome function
Antigen loading in exosomes:
LAMP2-mediated routing of antigens into exosomes in dendritic cells
Quantitative assessment of antigen enrichment in exosomal fractions
Functional analysis of LAMP2-rich exosomes in immune responses
Research has shown that KLH-pulsed monocyte-derived dendritic cells produce exosomes containing significantly more HLA-DR and KLH than control exosomes, and these exosomes are uniquely effective sources of antigen in T cell proliferation assays . This represents a novel pathway where LAMP2 directs antigens away from surface presentation and into highly immunogenic extracellular vesicles.
Anti-LAMP2 autoantibodies have been identified in certain autoimmune conditions, though their prevalence and pathogenic role remain subjects of ongoing research:
ANCA-associated vasculitis connection:
Some studies reported that >90% of patients with active pauci-immune glomerulonephritis had circulating anti-LAMP2 autoantibodies, though other research found lower frequencies
Many patients with anti-LAMP2 autoantibodies also have MPO-ANCA and PR3-ANCA
Various detection methods (ELISA, Western blot, immunofluorescence) show differing rates of positivity
Molecular mimicry hypothesis:
Anti-LAMP2 antibodies may recognize a nine-amino-acid peptide in bacterial adhesion protein (FimH) from fimbriated gram-negative bacteria, including Escherichia coli
Immunological response triggered by bacterial infection might lead to production of autoantibodies to human LAMP2
Animal studies showed that rats immunized with FimH peptide developed pauci-immune glomerulonephritis and antibodies to human LAMP2
Pathogenic potential:
Anti-LAMP2 autoantibodies have been proposed to be pathogenic in glomerulonephritis
They may disrupt lysosomal function and autophagy processes
If bacterial triggers are confirmed, this could have significant therapeutic implications
Diagnostic considerations:
Healthy individuals with active urinary tract infections may have antibodies reactive against recombinant LAMP2 at frequencies similar to ANCA-positive patients
Various assay systems show differing sensitivities and specificities
Multiple testing methodologies may be required for accurate assessment
The study of anti-LAMP2 autoantibodies represents an emerging field with potential implications for understanding the pathogenesis and treatment of certain autoimmune conditions.