EMC4 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Composition

EMC4 Antibody, FITC conjugated is a polyclonal antibody raised against a peptide sequence (amino acids 43–60) of the human EMC4 protein, covalently linked to FITC. EMC4 is a subunit of the ER membrane protein complex, critical for membrane protein biogenesis . FITC, a green-emitting fluorophore (λex=495nm,λem=525nm\lambda_{\text{ex}} = 495 \, \text{nm}, \lambda_{\text{em}} = 525 \, \text{nm}), enables fluorescence-based detection .

FITC Conjugation Chemistry

FITC binds to primary amine groups (lysine residues) on the antibody, forming stable thiourea linkages . Optimal conjugation requires:

  • pH: 9.5

  • Temperature: Room temperature (20–25°C)

  • Reaction Time: 30–60 minutes

Critical Parameters:

ParameterOptimal RangeImpact of Deviation
Fluorophore:Protein (F:P) Ratio3–6 FITC molecules per antibody Over-conjugation (>8 FITC/antibody) causes quenching and reduced binding affinity
Protein Concentration25 mg/mL Lower concentrations delay reaction kinetics

Primary Uses:

  1. Western Blot (WB): Detects EMC4 at 1:100–1:500 dilution .

  2. Immunohistochemistry (IHC): Stains formalin-fixed tissues at 1:10–1:50 dilution .

  3. Enzyme-Linked Immunosorbent Assay (ELISA): Sensitivity enhanced at 1:1000 dilution .

  4. Flow Cytometry: Enables live-cell analysis with minimal background .

Performance Data:

ApplicationRecommended DilutionSignal-to-Noise Ratio
Western Blot1:100–1:500>10:1
Immunofluorescence1:50>15:1

Research Findings and Validation

  • Binding Affinity: FITC conjugation reduces functional antibody concentration at high F:P ratios. For EMC4 antibodies, a 20–30% loss in avidity occurs at F:P > 6 .

  • Specificity: FITC-EMC4 shows no cross-reactivity with murine or rat homologs .

  • Staining Artifacts: Over-conjugated batches (>8 FITC/antibody) exhibit nonspecific binding in tissue sections .

Critical Assessments:

  • Kinetic ELISA: Quantifies active antibody fraction post-conjugation .

  • Fluorescence Quenching Assays: Monitor FITC-antibody stability under storage .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timelines.
Synonyms
EMC4; TMEM85; HSPC184; PIG17; ER membrane protein complex subunit 4; Cell proliferation-inducing gene 17 protein; Transmembrane protein 85
Target Names
EMC4
Uniprot No.

Target Background

Function
EMC4, a component of the endoplasmic reticulum (ER) membrane protein complex (EMC), facilitates the energy-independent insertion of newly synthesized membrane proteins into the ER membrane. This complex preferentially accommodates proteins with transmembrane domains that exhibit weak hydrophobicity or contain destabilizing features such as charged and aromatic residues. EMC4 plays a crucial role in the cotranslational insertion of multi-pass membrane proteins, where stop-transfer membrane-anchor sequences become ER membrane-spanning helices. Additionally, it is essential for the post-translational insertion of tail-anchored (TA) proteins into ER membranes. By mediating the proper cotranslational insertion of N-terminal transmembrane domains in an N-exo topology (with the translocated N-terminus in the lumen of the ER), EMC4 regulates the topology of multi-pass membrane proteins, such as G protein-coupled receptors. Through its regulation of protein insertion into membranes, EMC4 indirectly influences various cellular processes.
Gene References Into Functions
  1. TMEM85 is a complex gene that encodes a novel conserved anti-apoptotic protein. PMID: 18586032
Database Links

HGNC: 28032

OMIM: 616245

KEGG: hsa:51234

STRING: 9606.ENSP00000267750

UniGene: Hs.250905

Protein Families
EMC4 family
Subcellular Location
Endoplasmic reticulum membrane; Multi-pass membrane protein.
Tissue Specificity
Isoform 1 is expressed in brain and heart. Isoform 2 is expressed in heart.

Q&A

What is EMC4 and why is it significant in cellular research?

EMC4 (ER membrane protein complex subunit 4) is a critical component of the endoplasmic reticulum membrane protein complex involved in various cellular processes. The significance of EMC4 stems from its role in ER-associated protein folding, membrane protein insertion, and cellular stress responses that are fundamental to understanding cellular homeostasis. In research contexts, EMC4 serves as an important marker for studying ER function, protein trafficking, and cellular response to various stimuli or disease conditions. The detection of EMC4 using fluorescently labeled antibodies enables researchers to visualize its cellular localization and quantify its expression levels under different experimental conditions. Research using EMC4 antibodies has contributed significantly to our understanding of ER-associated degradation pathways and membrane protein quality control systems that are implicated in numerous diseases .

What are the key properties of FITC-conjugated antibodies?

FITC (Fluorescein Isothiocyanate) is a widely used fluorochrome with excitation and emission wavelengths of 499 nm and 515 nm, respectively, making it compatible with the 488 nm laser line commonly available in flow cytometers and fluorescence microscopes . The conjugation of FITC to antibodies creates a stable thiourea bond with primary amines of proteins, particularly lysine residues, allowing for sensitive detection of target antigens. FITC-conjugated antibodies exhibit bright green fluorescence when excited with the appropriate wavelength of light, providing excellent signal-to-noise ratio in fluorescence-based applications. The quantum yield of FITC is relatively high, contributing to its popularity in immunofluorescence techniques despite some susceptibility to photobleaching. The fluorescence intensity of FITC is pH-dependent, with optimal brightness occurring at alkaline pH (around 8-9), which researchers should consider when designing experiments and choosing appropriate buffers for sample preparation and analysis .

How should flow cytometry data for EMC4-FITC antibody be properly analyzed and interpreted?

Proper analysis of flow cytometry data for EMC4-FITC antibody begins with establishing appropriate gating strategies based on forward and side scatter properties to exclude cell debris and identify target cell populations. The fluorescence intensity data should be displayed as histograms for single-parameter analysis or as scatter plots when combined with other markers. When interpreting EMC4-FITC signals, it is essential to include proper controls such as isotype-matched FITC-conjugated antibodies to establish background fluorescence levels and determine positive signal thresholds. Quantitative analysis can be performed by calculating relative fluorescence intensity, which is the ratio between the fluorescence intensity of EMC4-FITC and the isotype control, allowing for normalized comparisons across different experiments or conditions. Researchers should be aware of potential fluorescence compensation requirements when using EMC4-FITC in combination with other fluorophores, particularly those with emission spectra that overlap with FITC, such as PE . For multiparameter analysis, scatter plots showing the expression of EMC4-FITC and another marker of interest provide more detailed information than single-parameter histograms, revealing cell subpopulations with distinct expression patterns that may be masked in histogram analysis alone .

What protocol is recommended for conjugating antibodies with FITC for custom applications?

The optimal protocol for FITC conjugation to antibodies involves several critical steps to ensure efficient labeling while preserving antibody functionality. First, purified antibody must be dialyzed against FITC labeling buffer (typically carbonate-bicarbonate buffer at pH 9.2) to remove free NH4+ ions and establish the alkaline conditions necessary for efficient conjugation. After determining the antibody concentration through absorbance measurements at 280 nm, researchers should add 20 μl of freshly prepared anhydrous FITC solution (5 mg/ml in DMSO) for each milligram of antibody and incubate the mixture for 2 hours at room temperature in the dark. Following conjugation, unbound FITC must be removed through dialysis against PBS or using gel filtration chromatography to prevent high background in subsequent applications. The fluorescein-to-protein (F/P) ratio should be determined spectrophotometrically, with optimal ratios typically between 2 and 5 for most applications, as higher ratios may lead to antibody inactivation or quenching effects. For maximum labeling efficiency, researchers should ensure high initial protein concentration (≥25 mg/ml), maintain alkaline pH (9.5), and control reaction temperature and time, with maximal labeling typically achieved within 30-60 minutes at room temperature under optimal conditions .

How can researchers optimize EMC4-FITC antibody dilutions for different applications?

Optimizing EMC4-FITC antibody dilutions requires a systematic approach tailored to each specific application. For flow cytometry, begin with the manufacturer's recommended range (typically 1:100-1:500) and perform a titration experiment using serial dilutions to identify the concentration that provides the highest signal-to-noise ratio. This optimization should include analysis of both staining index (difference between positive and negative populations divided by twice the standard deviation of the negative population) and separation index (difference between median fluorescence intensity of positive and negative populations). For fluorescence microscopy, prepare a dilution series starting from more concentrated preparations (1:10-1:50) and evaluate signal intensity, specificity, and background across multiple fields of view and biological replicates. Western blot applications typically require dilutions in the 1:100-1:500 range, but optimal concentration should be determined by testing multiple dilutions against known positive and negative control samples while monitoring both specific band intensity and background. For FLISA applications, where more dilute preparations (1:1000) are typically recommended, researchers should optimize by testing antibody concentrations against a standard curve of the target protein to determine the linear detection range and lower limit of detection . Throughout the optimization process, it is crucial to maintain consistent experimental conditions including incubation time, temperature, washing protocols, and detection settings to ensure reproducible results across experiments.

What are common troubleshooting strategies for weak or non-specific EMC4-FITC signals?

When encountering weak EMC4-FITC signals, several methodological adjustments can improve detection sensitivity. First, verify antibody integrity by checking for signs of degradation such as precipitation or unusual color changes, and consider testing a fresh aliquot if the antibody has undergone multiple freeze-thaw cycles. Increase antibody concentration incrementally while monitoring signal-to-noise ratio, as overconcentration can paradoxically reduce specificity due to increased non-specific binding. For fixed samples, optimize fixation protocols as overfixation can mask epitopes through protein cross-linking, while insufficient fixation may result in antigen loss during processing. For non-specific signals, implement more stringent blocking protocols using 5-10% normal serum from the same species as the secondary antibody (if used) or BSA in combination with Tween-20 or Triton X-100 to reduce hydrophobic interactions. Consider incorporating additional washing steps with higher salt concentration or detergent to remove weakly bound antibodies. If background persists, pre-adsorption of the antibody with non-target tissues or cells may reduce cross-reactivity. For flow cytometry applications specifically, ensure proper compensation when using multiple fluorophores to correct for spectral overlap, and include FcR blocking reagents when working with samples containing cells with Fc receptors that could bind antibodies non-specifically . Additionally, always compare results with appropriate positive and negative controls to distinguish between specific signals and experimental artifacts.

How should researchers store and handle EMC4-FITC antibody to maintain optimal activity?

Proper storage and handling of EMC4-FITC antibody is critical for maintaining its functionality and fluorescence properties over time. The antibody should be stored in small aliquots at -20°C in a non-frost-free freezer to prevent degradation from repeated freeze-thaw cycles, with each aliquot containing sufficient antibody for a single experiment to minimize waste and prevent contamination. When working with the antibody, always keep it on ice and protected from light to minimize photobleaching of the FITC fluorophore, which can significantly reduce signal intensity. For short-term storage (up to one month), the antibody can be kept at 4°C in the dark, but prolonged storage at this temperature is not recommended as it may lead to gradual loss of activity. The storage buffer typically contains 50% glycerol and preservatives like Proclin-300 to stabilize the antibody; do not dilute stock solutions unless immediately using them for experiments. Prior to each use, centrifuge the antibody vial briefly to collect the solution at the bottom of the tube and mix gently by pipetting rather than vortexing, which can denature the protein structure. Always check for signs of degradation such as precipitates or significant color changes before use, and if observed, the antibody should not be used for critical experiments . Importantly, EMC4-FITC antibody should never be frozen at temperatures below -20°C as this can compromise both antibody structure and fluorophore activity.

How can EMC4-FITC antibody be utilized in multi-parameter flow cytometry studies?

Multi-parameter flow cytometry with EMC4-FITC antibody enables sophisticated analysis of cellular heterogeneity by simultaneously examining EMC4 expression in conjunction with other cellular markers. When designing such panels, researchers must carefully select compatible fluorophores with minimal spectral overlap with FITC (excitation: 499 nm, emission: 515 nm) to reduce compensation requirements and prevent false positives. Typical compatible fluorophores include APC (excitation: ~650 nm, emission: ~660 nm) for red channel detection, PE-Cy7 for far-red detection, and Pacific Blue for violet channel detection. For experimental design, researchers should incorporate appropriate single-color controls for each fluorophore to establish compensation matrices that correct for spectral overlap. Analysis of multi-parameter data typically employs bivariate scatter plots to identify cellular subpopulations based on differential marker expression, as shown in flow cytometry studies where CD3 and CD4 expression were simultaneously assessed using FITC and APC conjugated antibodies, respectively . Advanced analytical approaches may include dimensionality reduction techniques such as tSNE or UMAP to visualize complex multi-parameter relationships, or clustering algorithms to identify novel cell populations based on marker expression patterns. The integration of EMC4-FITC detection with functional assays, such as intracellular cytokine staining or phospho-flow, can provide insights into how EMC4 expression correlates with cellular activation states or signaling pathway engagement in response to various stimuli.

What methodologies exist for using EMC4-FITC antibody in studying the ER membrane protein complex in disease models?

Advanced methodologies for applying EMC4-FITC antibody in disease model studies include several sophisticated approaches. Live-cell imaging combined with FRAP (Fluorescence Recovery After Photobleaching) can assess EMC4 dynamics within the ER membrane in real-time under various pathological conditions, providing insights into how disease states affect protein mobility and membrane distribution. Correlative light and electron microscopy (CLEM) techniques enable researchers to visualize EMC4 localization with fluorescence microscopy followed by ultrastructural analysis of the same cellular regions, offering unprecedented resolution of EMC4-containing membrane complexes in disease models. For animal studies, intravital microscopy using EMC4-FITC antibody allows for in vivo tracking of ER membrane complex dynamics in disease progression within intact tissues. Quantitative image analysis approaches such as co-localization studies with other ER markers (e.g., calnexin, KDEL receptors) can reveal alterations in ER organization and compartmentalization in disease states. Flow cytometry applications can be extended to patient-derived samples to identify changes in EMC4 expression levels across different cell populations in various diseases, particularly those affecting ER function such as neurodegenerative disorders or certain cancers. Integration with proteomics approaches, where EMC4-containing complexes are immunoprecipitated following FITC-antibody labeling and analyzed by mass spectrometry, can identify disease-specific interaction partners or post-translational modifications affecting EMC complex function . These methodologies collectively provide multidimensional insights into how EMC4 and the broader ER membrane protein complex may contribute to disease pathogenesis or adaptation.

How can researchers develop innovative applications using pH-dependent properties of FITC in EMC4 detection?

The pH-sensitivity of FITC conjugates offers exciting opportunities for developing innovative applications in EMC4 research. FITC fluorescence intensity increases significantly at alkaline pH (optimal at pH 8-9) and decreases in acidic environments, a property that can be leveraged to create pH-responsive EMC4 detection systems. Researchers can design protocols that exploit the inherent acidity of cellular compartments or pathological microenvironments, such as tumor tissues, which typically exhibit lower pH than normal tissues. This approach has been demonstrated with other FITC-conjugates that selectively bind to cancer cells in acidic tumor microenvironments, enabling targeted recruitment of antibodies for immune engagement . For EMC4 research, similar strategies could involve developing pH-sensitive EMC4-FITC probes that selectively label cells with altered ER pH, which can occur during ER stress responses or in certain disease states. These probes could be combined with ratiometric imaging techniques that normalize FITC signals against pH-insensitive fluorophores to generate quantitative maps of both EMC4 distribution and local pH environments. Advanced applications could include the development of EMC4-targeting theranostic agents where FITC serves both as an imaging reporter and as an antigenic epitope for therapeutic antibody recruitment, similar to approaches where FITC-decorated cancer cells induced humoral and cellular immunity against the fluorophore . Additionally, researchers could design EMC4-FITC conjugates with pH-cleavable linkers that release therapeutic payloads specifically in acidic organelles or disease microenvironments, creating targeted delivery systems for experimental therapeutics.

How does EMC4-FITC antibody compare with other detection methods for studying ER membrane proteins?

EMC4-FITC antibody offers distinct advantages over alternative detection methods for studying ER membrane proteins. Compared to indirect immunofluorescence techniques that require primary and secondary antibody incubations, directly conjugated EMC4-FITC antibodies streamline protocols by eliminating secondary antibody steps, reducing experimental time and potential cross-reactivity issues. When contrasted with genetically encoded fluorescent fusion proteins (e.g., EMC4-GFP), EMC4-FITC antibodies allow detection of endogenous protein without overexpression artifacts that may disrupt normal stoichiometry of the ER membrane complex or affect proper protein localization. EMC4-FITC detection offers superior specificity compared to small molecule ER dyes (e.g., ER-Tracker) that label the entire organelle rather than specific protein components. For quantitative analyses, EMC4-FITC antibodies in flow cytometry applications provide population-level data on expression heterogeneity that may be missed in traditional Western blot techniques, though Western blots offer advantages for distinguishing specific protein sizes to confirm target identity . The polyclonal nature of available EMC4-FITC antibodies provides signal amplification by recognizing multiple epitopes, potentially offering higher sensitivity than monoclonal alternatives, though at the cost of potential increased background and batch-to-batch variation . These comparisons highlight the importance of selecting detection methods based on specific experimental questions and technical considerations, with EMC4-FITC antibodies being particularly valuable for applications requiring single-step detection of endogenous protein in complex cellular systems.

What are emerging research directions for EMC4 antibodies in complement and immunity studies?

Emerging research directions for EMC4 antibodies intersect with complement and immunity studies in several innovative ways. Recent findings in complement biology, particularly studies using fluorescently labeled antibodies like anti-C4d FITC to detect complement activation during transplant rejection, provide methodological frameworks that could be applied to investigate whether EMC4 plays roles in complement-mediated cellular processes . The established protocols for detecting complement-activating anti-HLA alloantibodies using FITC-labeled antibodies could be adapted to explore potential relationships between EMC4 expression and complement cascade regulation, particularly in contexts of ER stress where unfolded protein responses may influence inflammatory signaling. Researchers are beginning to investigate whether EMC4 expression changes during immune cell activation or differentiation could serve as biomarkers for certain immunological conditions. The potential development of bispecific antibody constructs combining EMC4 recognition with immune effector recruitment represents another promising direction, drawing inspiration from studies where antigenic epitopes were used to selectively graft cancer cells for immune recognition . The role of EMC4 in antigen presentation machinery, particularly in professional antigen-presenting cells where ER quality control is essential for MHC loading, remains largely unexplored but could provide valuable insights into immunological processes. Additionally, investigating whether autoantibodies against EMC4 exist in certain autoimmune conditions affecting ER function could establish new diagnostic biomarkers, using methodologies similar to those employed in complement-dependent cytotoxicity (CDC) testing .

What future technical innovations might enhance the utility of FITC-conjugated antibodies in EMC4 research?

Future technical innovations in FITC-conjugated antibody technology will likely expand the capabilities of EMC4 research through several emerging approaches. Development of photoactivatable or photoswitchable FITC variants could enable super-resolution microscopy applications with EMC4 antibodies, allowing visualization of nanoscale ER membrane complex organization beyond the diffraction limit. Integration with microfluidic platforms could facilitate high-throughput screening of EMC4 expression across thousands of single cells under various treatment conditions, providing unprecedented insights into cellular heterogeneity in response to ER stressors. Advanced site-specific conjugation technologies that control the precise location and number of FITC molecules per antibody could enhance consistency and performance compared to current random conjugation methods that target all available lysine residues . Development of FITC-conjugated nanobodies or small recombinant antibody fragments against EMC4 would enable better penetration into complex samples and reduce the size differential between probe and target, potentially improving spatial resolution in imaging applications. Creation of dual-labeled antibodies combining FITC with long-wavelength fluorophores could enable FRET-based assays to detect conformational changes in EMC4 or its proximity to other ER membrane proteins under different cellular conditions. Implementation of DNA-barcoded EMC4-FITC antibodies compatible with spatial transcriptomics techniques would allow simultaneous visualization and molecular identification of EMC4-expressing cells in complex tissues. These innovations reflect the convergence of antibody engineering, fluorophore chemistry, and advanced imaging technologies that will collectively enhance our ability to study EMC4 biology with unprecedented precision and contextual information.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.