The MPEG1 antibody is a polyclonal or monoclonal immunoglobulin that binds specifically to the MPEG1 protein. It is used to:
Detect protein expression: In techniques like Western blotting (WB) and immunofluorescence (IF).
Study localization: Through immunohistochemistry (IHC) and immunocytochemistry (ICC).
Investigate interactions: Via immunoprecipitation (IP).
MPEG1 antibodies are validated for human, mouse, or rat samples, depending on the product. Key applications include studying bacterial resistance, immune regulation, and chronic inflammation mechanisms .
MPEG1 antibodies are utilized across diverse experimental workflows:
Bacterial resistance: MPEG1-deficient mice and zebrafish show heightened susceptibility to Mycobacterium marinum and Staphylococcus aureus .
Pore-forming activity: Recombinant MPEG1 disrupts bacterial membranes, aiding antimicrobial effector entry .
Chronic inflammation: Aged MPEG1-deficient mice exhibit microbial translocation from the gut, leading to sustained B-cell inflammation and reduced antibody responses .
IFN signaling: MPEG1 regulates interferon-α/β pathways, linking bacterial burden to autoimmune disease risk .
MPEG1 is an ancient metazoan protein belonging to the MACPF/CDC superfamily of pore-forming proteins that plays a crucial role in innate immunity. Its significance stems from several key aspects: it represents an evolutionary ancestor to better-known immune effectors like complement component C9 and perforin; it functions as an intracellular pore-forming immune effector; and its expression is upregulated in response to proinflammatory signals such as TNFα and lipopolysaccharides (LPS) . Studies have demonstrated that partial or complete loss of MPEG1 causes increased susceptibility to microbial infection in both cells and animal models, highlighting its importance in antimicrobial defense . Furthermore, germline mutations in MPEG1 have been identified in connection with recurrent pulmonary mycobacterial infections in humans, underscoring its clinical relevance . Research examining MPEG1 can provide insights into fundamental mechanisms of innate immunity and potentially inform therapeutic approaches for infectious diseases.
MPEG1 expression is dynamically regulated by several proinflammatory signals. For researchers designing stimulation experiments, the following protocols have been validated: TNFα and LPS independently drive MPEG1 expression through MyD88 and NFκB pathways, with additive effects when used in combination . While IFNγ alone has minimal effect on MPEG1 expression, it synergistically enhances expression when combined with LPS . When studying the short isoform MPEG1b, which lacks the transmembrane domain, note that LPS stimulation not only increases expression but also enhances secretion . For optimal results when inducing MPEG1 expression, researchers should employ time-course experiments, typically ranging from 6 to 24 hours of stimulation, to capture peak expression levels. The activation state of macrophages significantly affects MPEG1 expression levels, so researchers should carefully document polarization conditions (M1 vs. M2) when interpreting antibody staining results. When developing stimulation protocols, consider cell-type specific responses, as primary cells and cell lines may differ in their MPEG1 induction kinetics and magnitude.
To investigate MPEG1's pore-forming function, researchers can employ multiple complementary approaches using specific antibodies. Immunofluorescence co-localization experiments with antibodies targeting both MPEG1 and bacterial membrane markers can reveal MPEG1 association with pathogen membranes during infection . For mechanistic studies, researchers should design time-course experiments using confocal microscopy to visualize the trafficking of MPEG1 to phagosomes containing pathogens. Importantly, antibodies recognizing different domains of MPEG1 (MACPF domain, MABP domain, cytoplasmic tail) can help distinguish between intact protein and processed fragments during pore formation . Immunoprecipitation combined with Western blotting can detect oligomeric complexes formed during pore assembly, particularly after limited proteolysis treatments . To directly visualize MPEG1 pores, researchers can isolate membranes from infected cells, perform immunogold labeling with MPEG1 antibodies, and analyze the samples using electron microscopy, which has successfully revealed ring-like oligomeric structures . When studying pH-dependent pore formation, researchers should employ antibodies that maintain reactivity under acidic conditions, as structural studies have shown that MPEG1's lytic activity is strictly dependent on low pH .
Proteolytic processing is critical for MPEG1 function, as evidenced by the detection of ectodomain fragments in bacterial membranes while the C-terminal cytosolic region remains in host membranes . To study this phenomenon, researchers should employ domain-specific antibodies that can distinguish between intact MPEG1 and its processed fragments. Western blotting using antibodies against different domains can track processing events by detecting size shifts. For identifying cleavage sites, immunoprecipitation with domain-specific antibodies followed by mass spectrometry analysis can precisely map the proteolytic fragments. Pulse-chase experiments combined with immunoprecipitation can establish the kinetics of MPEG1 processing during infection or stimulation. To investigate the functional significance of proteolysis, researchers can generate cleavage-resistant MPEG1 mutants and analyze their pore-forming capacity using antibodies in oligomerization assays . For in situ visualization of processing events, proximity ligation assays using antibodies against different MPEG1 domains can reveal spatial and temporal aspects of proteolysis. Although the endogenous protease responsible for MPEG1 cleavage remains unidentified, protease inhibitor panels combined with MPEG1 antibody detection can help narrow down candidate proteases .
Recent research has implicated MPEG1 in regulating type I interferon signaling through its interactions with the Interferon-α/β receptor (IFNAR) complex . To investigate this emerging function, researchers should design co-immunoprecipitation experiments using antibodies against MPEG1 and IFNAR components to confirm their physical association. Proximity ligation assays with antibodies against MPEG1 and IFNAR1/IFNAR2 can visualize their interactions in situ. Phospho-specific antibodies against downstream signaling components (STAT1, STAT2, JAK1, TYK2) should be employed to assess the impact of MPEG1 knockdown or knockout on signaling pathway activation . Domain-mapping studies using truncated MPEG1 constructs and domain-specific antibodies can help identify which regions of MPEG1 (MACPF domain, MABP domain, cytosolic region) mediate the interactions with IFNAR components, as previous studies suggest the MACPF and MABP domains interact with IFNAR1 and IFNAR2 respectively, while the cytosolic region is required for STAT2 phosphorylation . For functional studies, researchers should monitor interferon-stimulated gene expression in MPEG1-deficient versus wild-type cells using both antibody-based techniques (Western blot, flow cytometry) and transcriptional analysis. When studying MPEG1's role in excessive IFN signaling during overwhelming immune responses, antibodies can track MPEG1 expression changes during LPS-induced septic shock models, where MPEG1-deficient mice show resistance .
When designing subcellular fractionation experiments to study MPEG1 localization and trafficking, researchers should implement a systematic approach that accounts for MPEG1's complex distribution pattern. Since MPEG1 traffics through the secretory pathway and localizes to several compartments (ER, Golgi, secretory vesicles, early endosomes, phagosomes/lysosomes, and plasma membrane), differential centrifugation followed by gradient separation provides optimal resolution . Begin with gentle lysis buffers containing protease inhibitors to preserve MPEG1 integrity, as proteolytic processing occurs naturally during MPEG1 function. For membrane-bound MPEG1, detergent solubilization conditions should be carefully optimized, as harsh detergents may disrupt MPEG1 oligomeric structures that form during pore assembly. When analyzing fractions by Western blotting, researchers should employ antibodies against both the ectodomain and cytosolic regions of MPEG1 to track potential proteolytic fragments . For validation, include antibodies against compartment-specific markers such as EEA1 (early endosomes), LAMP1 (lysosomes), and organelle markers for the ER and Golgi. In stimulated conditions, particularly after infection or inflammatory activation, expect altered distribution patterns as MPEG1 redistributes to pathogen-containing compartments. When studying secreted MPEG1b (the short isoform lacking the transmembrane domain), concentrate culture supernatants before antibody detection, as this form is released into the extracellular space .
Rigorous controls and validation are critical for reliable MPEG1 antibody experiments. For positive controls, use cells known to constitutively express MPEG1 (macrophages, leukocytes) or stimulated epithelial/fibroblast cells following TNFα/LPS treatment . Essential negative controls include MPEG1 knockout/knockdown cells, which should be generated using CRISPR-Cas9 or RNAi technologies to confirm antibody specificity. When multiple MPEG1 paralogs exist (as in zebrafish), paralog-specific antibodies should be validated using individual knockout models for each paralog . For peptide competition assays, pre-incubate the antibody with excess immunizing peptide to confirm binding specificity. Cross-species reactivity should be carefully assessed, as MPEG1 sequence conservation varies across evolutionary lineages. When studying MPEG1 in tissue sections, include isotype controls matched to the MPEG1 antibody to distinguish specific staining from background. For stimulation experiments, parallel samples with and without cytokine/LPS treatment serve as internal controls for antibody performance . When investigating proteolytic processing, compare antibodies targeting different MPEG1 domains to confirm fragment identity. Finally, verify antibody specificity through orthogonal methods, such as correlating protein detection with mRNA expression using RT-PCR or RNA-seq data.
When designing infection experiments to study MPEG1's antimicrobial function, researchers should implement comprehensive approaches that capture its dynamic behavior. For in vitro infection models, synchronize infection by centrifuging bacteria onto cell monolayers, and perform time-course analyses using MPEG1 antibodies to track recruitment to pathogen-containing phagosomes . Confocal microscopy with Z-stack acquisition is essential for accurately visualizing MPEG1 colocalization with intracellular pathogens. When studying bacterial killing mechanisms, compare wild-type cells with MPEG1-deficient counterparts, as multiple studies have demonstrated increased bacterial survival in MPEG1 knockout models . For mechanistic insights into MPEG1-mediated killing, combine antibody detection with bacterial viability assays and membrane integrity tests. In mycobacterial infection models, particularly relevant given MPEG1's connection to mycobacterial infections in humans, monitor bacterial cell wall integrity after MPEG1 recruitment, as previous studies have observed mycobacterial swelling following MPEG1 treatment . For in vivo infection studies, tissue-specific immunohistochemistry using MPEG1 antibodies can reveal infection-induced expression changes across different organs. When analyzing MPEG1's role in facilitating entry of other antimicrobial effectors into bacteria, design co-localization experiments with antibodies against MPEG1 and various antimicrobial proteins (proteases, bactericidal peptides) . Finally, to study pH-dependent activity, use pH-sensitive fluorescent probes in conjunction with MPEG1 antibody staining to correlate phagosomal acidification with MPEG1 function .
Researchers frequently encounter several technical challenges when working with MPEG1 antibodies. One common issue is weak or absent staining despite expected MPEG1 expression, which may result from insufficient upregulation in the experimental conditions used. To address this, consider extending stimulation times with TNFα and LPS, as MPEG1 expression is significantly enhanced by these proinflammatory signals . If detecting the short isoform MPEG1b, remember it lacks the transmembrane domain and is secreted, requiring analysis of culture supernatants rather than cell lysates . Another challenge is distinguishing specific MPEG1 staining from background in tissues with high autofluorescence. In this case, implement spectral unmixing techniques and include appropriate isotype controls. Inconsistent subcellular localization patterns may occur due to MPEG1's dynamic trafficking between compartments (ER, Golgi, endosomes, phagosomes, plasma membrane) . To address this, synchronize cells before fixation and use compartment-specific markers for co-localization. When studying MPEG1 proteolytic processing, antibodies against different domains may yield conflicting results; this actually reflects the biological reality of domain separation during function . For optimal detection of membrane-associated MPEG1, mild permeabilization protocols are recommended to preserve membrane structure while allowing antibody access. If studying MPEG1's pore-forming activity, note that structural changes during oligomerization may mask epitopes, requiring different antibodies for detection of monomeric versus oligomeric forms .
Interpreting MPEG1 expression patterns requires careful consideration of multiple biological variables. First, recognize that cell type-specific expression is normal; constitutive expression occurs primarily in macrophages and leukocytes, while other cell types (epithelial, fibroblast) require stimulation for expression . Differential expression between in vitro and in vivo samples is common due to the complex cytokine milieu in living organisms versus defined culture conditions. When analyzing MPEG1 expression in response to pathogens, consider pathogen-specific effects; different infectious agents may induce unique expression patterns or timing. For instance, in zebrafish, different MPEG1 paralogs respond distinctly to Mycobacterium marinum infection (mpeg1.2 upregulation versus mpeg1 suppression) . The kinetics of expression also vary with stimulation type; combined TNFα/LPS treatment produces additive effects, while IFNγ alone has minimal impact but synergizes with LPS . When examining subcellular distribution, interpret changes in pattern as potentially functional; redistribution to pathogen-containing compartments suggests active involvement in antimicrobial defense. In tissues from infection models, heterogeneous MPEG1 expression likely reflects local differences in inflammatory signaling. For quantitative comparisons across conditions, normalize MPEG1 levels to appropriate housekeeping proteins and cell-type specific markers. Finally, consider the potential impact of genetic background, as polymorphisms affecting MPEG1 expression or function may exist in research models or human samples .
When quantifying MPEG1 expression using antibody-based methods, researchers should implement robust statistical approaches that account for biological variability and technical limitations. For Western blot quantification, perform at least three independent biological replicates and normalize MPEG1 signals to loading controls appropriate for the subcellular fraction being analyzed; total protein normalization (using stain-free gels or Ponceau staining) is preferable to single housekeeping proteins, which may vary under stimulation conditions . When comparing MPEG1 expression across multiple stimulation conditions (TNFα, LPS, IFNγ, combinations), use ANOVA with appropriate post-hoc tests rather than multiple t-tests to control for family-wise error rates . For immunofluorescence quantification, analyze multiple fields per condition (>10) and multiple cells per field (>50) to account for heterogeneous expression. Z-score normalization can help compare expression across different experiments when absolute values cannot be directly compared. When conducting flow cytometry analysis of MPEG1 expression, include fluorescence-minus-one (FMO) controls to set proper gating strategies. For time-course experiments, consider area-under-the-curve analyses rather than single timepoint comparisons to capture expression dynamics. To validate antibody-based quantification, correlate results with orthogonal methods such as RT-qPCR for mRNA levels or mass spectrometry for protein levels. Finally, when comparing MPEG1 expression between wild-type and disease models (e.g., infection models), use matched controls and consider potential confounding variables such as changes in cellular composition or activation state .
The evolutionary relationship between MPEG1 and other MACPF/CDC superfamily members offers fascinating research opportunities using comparative antibody-based approaches. Researchers can design epitope mapping studies using antibodies against conserved regions of MPEG1, perforin, and complement components to identify structural elements maintained throughout evolution . Cross-reactivity analyses with antibodies raised against MPEG1 from different species (sponge, oyster, fish, mouse, human) can reveal evolutionary conservation of functional domains . To investigate the hypothesis that gene duplications of MPEG1 gave rise to perforin and MAC components, researchers can use antibodies in comparative localization studies across species representing different evolutionary time points. Domain-specific antibodies targeting the MACPF domain versus other regions can help determine which structural elements are most conserved. For functional comparative studies, researchers can investigate whether antibodies against MPEG1 from ancient organisms (like sponges) recognize mammalian MPEG1, providing insights into structural conservation across >700 million years of evolution . Comparative immunoprecipitation studies using MPEG1 antibodies followed by mass spectrometry can identify interacting protein networks across species, revealing conservation or divergence of functional pathways. When studying pore formation mechanisms, researchers should compare oligomeric structures formed by MPEG1 versus other MACPF proteins using antibody-based visualization techniques, as MPEG1's unusual domain arrangement suggests a novel mechanism of pore formation that may have evolved to guard against unwanted lysis of the host cell .
MPEG1's role in immune regulation makes it a promising target for investigating chronic inflammation and autoimmune conditions. Recent studies with aged MPEG1-deficient mice revealed increased microbial migration from the gastrointestinal tract into serum, resulting in chronic inflammation and altered B cell responses . To explore similar mechanisms in human conditions, researchers can use MPEG1 antibodies for immunohistochemical analysis of tissues from patients with inflammatory bowel diseases, comparing expression patterns with bacterial localization and inflammatory markers. For investigating MPEG1's potential role in barrier function, combine antibody staining with markers of epithelial integrity in experimental colitis models. Since MPEG1 regulates type I interferon signaling, a pathway implicated in multiple autoimmune diseases, researchers should analyze MPEG1 expression and localization in tissues from patients with systemic lupus erythematosus, Sjögren's syndrome, and other interferon-driven conditions . To study how MPEG1 might influence B cell abnormalities in autoimmunity, design co-culture experiments with MPEG1-sufficient or deficient macrophages and B cells, using antibodies to track changes in B cell activation markers . For mechanistic studies of MPEG1 in chronic inflammation, use flow cytometry with MPEG1 antibodies to quantify expression in different immune cell populations from peripheral blood or affected tissues of patients versus healthy controls. Time-course studies during experimental autoimmune disease induction can reveal whether MPEG1 expression changes precede or follow inflammatory cascades. Finally, given MPEG1's role in LPS-induced shock resistance in knockout models, researchers should investigate whether MPEG1 antibody blockade could modulate excessive inflammatory responses in sepsis models .
Given the connection between MPEG1 germline mutations and recurrent pulmonary mycobacterial infections in humans, specialized approaches for mycobacterial research are warranted . Researchers should design infection models with clinically relevant mycobacterial species (M. tuberculosis, M. avium, M. abscessus) in both wild-type and MPEG1-deficient cells, using antibodies to track MPEG1 recruitment to mycobacterial phagosomes. Super-resolution microscopy with dual-labeled antibodies can visualize the spatial relationship between MPEG1 and the mycobacterial cell wall during different infection stages. To investigate the mechanism of MPEG1 activity against mycobacteria, combine antibody localization with assays measuring mycobacterial membrane integrity, as previous studies observed mycobacterial swelling after MPEG1 treatment . Time-lapse imaging with fluorescently labeled antibodies (Fab fragments) in live infected cells can capture the dynamics of MPEG1 recruitment and potential mycobacterial responses. To explore MPEG1's role in facilitating entry of other antimicrobial effectors, design co-localization experiments with antibodies against MPEG1, lysozyme, and other antimicrobial proteins, as studies suggest MPEG1 perforates the outer membrane while other factors attack the peptidoglycan layer . For translational research, analyze MPEG1 expression in bronchoalveolar lavage samples or lung biopsies from patients with mycobacterial infections versus healthy controls. When studying MPEG1 variants identified in patients with recurrent mycobacterial infections, use structure-informed antibodies that can distinguish between wild-type and mutant conformations to assess potential functional differences. Finally, in animal models of mycobacterial infection, tissue-specific immunohistochemistry with MPEG1 antibodies can map expression patterns in relation to granuloma formation and bacterial containment .