The REM1 Antibody, Biotin conjugated, is a research-grade polyclonal antibody designed for detecting the REM1 (RAS (RAD and GEM)-Like GTP-Binding 1) protein. REM1, a 33 kDa GTP-binding protein, plays roles in endothelial cell sprouting, actin cytoskeletal reorganization, and angiogenesis . The biotin conjugation enhances detection sensitivity by leveraging the high-affinity biotin-streptavidin interaction, enabling applications in ELISA, Western blotting, and immunohistochemistry (IHC) .
The biotin conjugate enables indirect detection via streptavidin-linked reporters (e.g., HRP, alkaline phosphatase, fluorophores) . Common applications include:
ELISA: Quantitative detection of REM1 in solution, paired with streptavidin-HRP or AP for signal amplification .
Western Blotting: Though not explicitly tested for the biotin-conjugated variant, biotinylated antibodies are often used here .
Immunoprecipitation: Potential use in isolating REM1-protein complexes using streptavidin-coated beads .
Signal Amplification: Biotin-streptavidin interactions (K<sub>d</sub> ~10<sup>−14</sup>–10<sup>−15</sup> M) enable high sensitivity .
Versatility: Compatible with multiple streptavidin-linked enzymes/fluorophores, allowing flexibility in assay design .
Reduced Background: Avoids direct enzyme conjugation, minimizing nonspecific binding .
Target Specificity: Detects endogenous REM1 protein in human samples .
Cross-Reactivity: Predicted reactivity with pig, bovine, horse, and sheep (based on sequence homology) .
Optimal Dilutions: Suggested ELISA dilution: 1:20,000–1:40,000 .
Restricted Applications: Primarily validated for ELISA; optimization required for other methods (e.g., IHC) .
Endogenous Biotin Interference: Mitochondrial biotin-containing proteins may cause background in IHC; blocking agents recommended .
| Conjugate Type | Advantages | Disadvantages |
|---|---|---|
| Biotin | High sensitivity, versatile detection | Requires streptavidin reagents |
| HRP | Direct detection, no additional steps | Lower sensitivity, limited flexibility |
| Fluorophores | Multiplexing capability | Photobleaching, lower signal stability |
REM1 (RAS (RAD and GEM)-Like GTP-binding 1) is a member of the RAS superfamily of small GTPases that functions as a molecular switch in various cellular signaling pathways. This protein is particularly interesting for researchers studying signal transduction, cellular proliferation, and potential cancer mechanisms. REM1 shares structural similarities with other RAS family proteins, containing characteristic GTP-binding domains that enable its participation in cellular signaling cascades. Its full-length human protein consists of 298 amino acids with distinct functional domains, including the highly conserved GTP-binding region that is critical for its biological activity . The antibodies against REM1 allow researchers to investigate the expression, localization, and function of this protein in various experimental contexts.
Commercial REM1 antibodies are designed to recognize different epitopes within the protein, offering researchers flexibility in experimental design. Available antibodies include those targeting the N-terminal region (amino acids 2-153), which contains important regulatory domains, as well as antibodies recognizing the full-length protein (amino acids 1-298) . The epitope selection is critically important as it determines which functional domains or protein states can be detected. For instance, antibodies targeting amino acids 2-153 may be particularly useful for studying the N-terminal regulatory regions of REM1, while those recognizing amino acids 221-298 might be more suitable for investigating C-terminal interactions. The immunogen sequence "MTLNTEQEAKTPLHRRASTPLPLSPRGHQPGRLSTVPSTQSQHPRLGQSASLNPPTQKPSPAPDDWSSESSDSEGSWEALYRVVLLGDPGVGKTSLASLFAGKQERDLHEQLGEDVYERTLTVDGEDTTLVVVDTWEAEKLDKSWSQESCLQGGSAYVIVYSIADRGSFESASELRIQLRRTHQADHVPIILVGNKADLARCREVSVEEGRACAVVFDCKFIETSATLQHNVAELFEGVVRQLRLRRRDSAAKEPPAPRRPASLAQRARRFLARLTARSARRRALKARSKSCHNLAVL" provides the basis for full-length REM1 antibody generation, containing the GTP-binding domain and other functional regions .
The choice of host species for antibody production significantly impacts research applications. REM1 antibodies are typically produced in either mouse or rabbit hosts, each with distinct advantages. Mouse-derived polyclonal antibodies against full-length REM1 (amino acids 1-298) typically demonstrate strong reactivity in Western blot and immunofluorescence applications with human samples . Rabbit-derived polyclonal antibodies targeting specific regions like amino acids 2-153 generally show excellent performance in ELISA and immunofluorescence applications . The host species influences the antibody's compatibility with other reagents in multiplexing experiments. For instance, when performing co-localization studies, selecting antibodies from different host species prevents cross-reactivity between secondary detection antibodies. Additionally, the host immune system's response to the immunogen affects the diversity of epitopes recognized, with rabbits typically generating antibodies with higher affinity and broader epitope recognition compared to mice for the same antigen.
Biotin conjugation to REM1 antibodies involves the covalent attachment of biotin molecules to the antibody structure through specific chemical reactions. The process typically targets primary amine groups (-NH2) on lysine residues or the N-terminus of the antibody protein. Modern conjugation kits like the LYNX Rapid Plus Biotin Type 1 system employ activated biotin derivatives that react spontaneously with antibody amines under mild conditions, forming stable amide bonds . The reaction conditions are carefully controlled to achieve optimal biotin:antibody ratios (typically 3-7 biotin molecules per antibody) while preserving the antigen-binding capacity. This precise control is essential because over-biotinylation can interfere with the antibody's binding region, while under-biotinylation results in reduced detection sensitivity. The conjugation process must maintain the antibody's native conformation and antigen-binding properties while introducing sufficient biotin molecules for downstream detection using streptavidin conjugates. The resulting biotin-conjugated REM1 antibodies retain their specificity for the original epitope (such as amino acids 2-153) while gaining the ability to interact with extraordinary affinity to streptavidin detection systems .
Biotin-conjugated REM1 antibodies enable multiple signal amplification strategies that can dramatically enhance detection sensitivity in various applications. The primary amplification mechanism leverages the tetravalent binding capacity of streptavidin, which can bind four biotin molecules simultaneously. This property allows researchers to implement multi-layer detection systems, such as:
Sequential amplification using biotinylated tyramine, which deposits additional biotin molecules near the primary binding site through a peroxidase-catalyzed reaction, providing a substantial increase in local biotin concentration.
Streptavidin-conjugated reporter enzymes (HRP or alkaline phosphatase) that catalyze chromogenic or chemiluminescent reactions, generating multiple detectable molecules per bound antibody.
Fluorescent amplification using streptavidin conjugated to quantum dots or fluorescent proteins, which provide brighter and more photostable signals than conventional fluorophores.
Pretargeted radioimmunotherapy (PRIT) strategies that use biotin-streptavidin interactions to deliver therapeutic radionuclides with improved tumor-to-normal tissue ratios compared to directly labeled antibodies .
These amplification techniques enable detection of REM1 proteins expressed at very low levels, which might be undetectable with conventional direct-labeled antibodies. For instance, in immunohistochemistry applications, biotin-streptavidin amplification can improve sensitivity by 4-8 fold compared to direct detection methods.
Optimizing Western blot protocols for biotin-conjugated REM1 antibodies requires careful consideration of several parameters to maximize specificity and sensitivity. Based on available research data, the following methodological approach is recommended:
Sample preparation: Begin with effective cell lysis using compatible buffers (typically PBS-based with protease inhibitors) to maintain the native structure of REM1 protein. For REM1 detection, transfected lysates have been successfully used as positive controls, showing a band at approximately 32.78 kDa .
Blocking: To minimize background signal, use casein-based blocking buffers rather than those containing biotin (such as milk) which can interfere with the biotin-streptavidin detection system.
Primary antibody incubation: Dilute the biotin-conjugated REM1 antibody to 1:500 in blocking buffer and incubate overnight at 4°C for optimal binding . This dilution has been validated for Western blot applications with REM1 antibodies.
Detection system: Employ streptavidin-HRP at a 1:2000-1:5000 dilution with a brief incubation (30-60 minutes) at room temperature. The high affinity of the biotin-streptavidin interaction allows for shorter incubation times compared to secondary antibody methods.
Signal development: Enhanced chemiluminescence (ECL) substrates are recommended, with exposure times typically ranging from 30 seconds to 5 minutes depending on REM1 expression levels.
Controls: Include non-transfected lysate controls alongside REM1-transfected samples to verify specificity, as demonstrated in published Western blot analyses .
This optimized protocol enables clear detection of REM1 protein bands with minimal background, providing reliable quantitative data for expression analysis.
Successful immunofluorescence studies using biotin-conjugated REM1 antibodies require meticulous attention to fixation methods, antigen retrieval, and detection strategies. The following methodological approach has been validated for REM1 localization studies:
Cell preparation: HeLa cells have been successfully used for REM1 immunofluorescence studies, with optimal cell density at 70-80% confluence to maintain proper morphology .
Fixation: Paraformaldehyde (4%) fixation for 15 minutes at room temperature preserves cellular architecture while maintaining REM1 epitope accessibility. Avoid methanol fixation which can denature GTP-binding proteins.
Antigen retrieval: A mild citrate buffer (pH 6.0) heat-induced epitope retrieval step may enhance detection of certain REM1 epitopes, particularly in tissue sections.
Antibody concentration: An optimal concentration of 10 μg/ml has been empirically determined for biotin-conjugated REM1 antibodies in immunofluorescence applications . This concentration provides sufficient signal while minimizing background.
Detection system: Fluorophore-conjugated streptavidin (Alexa Fluor 488, 555, or 647) provides excellent sensitivity and spectral compatibility for co-localization studies.
Nuclear counterstaining: DAPI at 300 nM provides good nuclear definition without interfering with REM1 signal detection.
Autofluorescence reduction: Treatment with 0.1% sodium borohydride before blocking can reduce cellular autofluorescence that might interfere with REM1 detection.
These validated protocols allow researchers to precisely visualize REM1 subcellular localization and study its potential co-localization with other proteins of interest in signaling pathways.
Developing robust quantitative ELISA assays with biotin-conjugated REM1 antibodies requires systematic optimization of multiple parameters to ensure sensitivity, specificity, and reproducibility. The following methodological approach is recommended based on research data:
Assay format selection: A sandwich ELISA using an unconjugated capture antibody (targeting one REM1 epitope) and a biotin-conjugated detection antibody (targeting a different epitope) provides optimal specificity. For instance, combining antibodies that recognize amino acids 2-153 and 221-298 regions can create a highly specific detection system .
Plate coating: Coat high-binding ELISA plates with capture antibody at 2-5 μg/ml in carbonate buffer (pH 9.6) overnight at 4°C, followed by thorough washing.
Blocking optimization: A 2% BSA solution in PBS effectively blocks non-specific binding without interfering with the biotin-streptavidin interaction.
Sample preparation: Cell lysates should be prepared in RIPA buffer with protease inhibitors, centrifuged at high speed (>10,000 g) to remove debris, and diluted appropriately in assay buffer.
Standard curve generation: Recombinant REM1 protein titrated from 0.1-100 ng/ml establishes a reference for quantification.
Detection system: Streptavidin-HRP diluted 1:5000-1:10000 provides sensitive detection without excessive background. TMB substrate with sulfuric acid stop solution offers good dynamic range.
Validation parameters:
Sensitivity: Typical detection limits range from 0.1-0.5 ng/ml of REM1 protein
Dynamic range: 0.5-50 ng/ml for reliable quantification
Precision: Intra-assay CV <10%, inter-assay CV <15%
Specificity: Confirmed through lack of signal with related GTPases
This methodologically rigorous approach enables accurate quantification of REM1 protein in research samples with high reproducibility.
Background signals represent a significant challenge when working with biotin-conjugated REM1 antibodies, stemming from multiple potential sources that require systematic troubleshooting. The following methodological analysis addresses the most common causes and their solutions:
Endogenous biotin interference: Mammalian cells naturally contain biotin, which can bind to streptavidin detection reagents. This issue is particularly pronounced in biotin-rich tissues like liver, kidney, and brain. Pre-blocking with unconjugated streptavidin (10-15 μg/ml) followed by free biotin (50-100 μg/ml) effectively masks endogenous biotin.
Non-specific antibody binding: Polyclonal REM1 antibodies may contain antibody populations that bind non-specifically to cellular components. Increasing blocking agent concentration (3-5% BSA or casein) and adding 0.1-0.2% Triton X-100 to washing buffers can reduce these interactions.
Over-concentration of detection reagents: Excessive concentration of streptavidin-conjugated reporters leads to increased non-specific binding. Careful titration starting from 1:2000 dilution of streptavidin-HRP or 1:500 of fluorescent streptavidin conjugates generally identifies optimal concentrations.
Sample-specific autofluorescence: In immunofluorescence applications, certain fixatives increase cellular autofluorescence. Treatment with 0.1% sodium borohydride or 50 mM NH4Cl after fixation reduces this background.
Biotin in blocking reagents: Milk-based blocking solutions contain endogenous biotin. Researchers should exclusively use BSA, casein, or commercial biotin-free blocking reagents.
Controlled experiments have demonstrated that implementing these strategic interventions can improve signal-to-noise ratios by 3-5 fold in immunoassays using biotin-conjugated REM1 antibodies.
Validating the specificity of biotin-conjugated REM1 antibodies requires a multi-faceted approach combining positive and negative controls with competitive binding assays. The following methodological strategy ensures reliable validation:
Comparative Western blotting: Analyze REM1-transfected versus non-transfected cell lysates. Specific antibodies should detect a band at approximately 32.78 kDa in transfected samples only, as demonstrated with validated REM1 antibodies .
Peptide competition assays: Pre-incubate the antibody with excess (10-100 μg/ml) of the immunizing peptide before application to samples. Specific binding should be significantly reduced or eliminated.
Knockdown verification: Analyze samples from cells with REM1 expression silenced through siRNA or CRISPR-Cas9. Signal reduction proportional to knockdown efficiency confirms specificity.
Cross-reactivity testing: Evaluate antibody reactivity against related GTPases (GEM, RAD) to ensure selective recognition of REM1.
Immunoprecipitation-mass spectrometry: Pull down proteins using the REM1 antibody and identify them by mass spectrometry. Specific antibodies predominantly enrich REM1 protein.
Flow cytometry competitive binding assays: Use fluorescein-labeled standard antibodies in competition with the biotin-conjugated antibody. Similar IC50 values (concentration required for 50% inhibition) indicate recognition of the same epitope .
Immunocytochemistry specificity controls: Include appropriate negative controls such as isotype-matched irrelevant antibodies and positive controls such as cells known to express REM1.
This systematic validation approach ensures that experimental observations accurately reflect REM1 biology rather than artifacts of non-specific antibody interactions.
Comprehensive quality control of biotin-conjugated REM1 antibodies requires evaluation of multiple critical parameters to ensure consistent performance across experiments. The following methodological approach addresses key quality metrics:
Biotinylation degree determination: The biotin:protein ratio should be quantified using HABA (4'-hydroxyazobenzene-2-carboxylic acid) assay or mass spectrometry. Optimal ratios typically range from 3-7 biotin molecules per antibody, balancing detection sensitivity with preservation of binding capacity.
Immunoreactivity assessment: Compare the antigen-binding capacity of biotinylated versus unconjugated antibody preparations. Well-conjugated antibodies maintain >80% of original immunoreactivity. This can be assessed through comparative ELISA or flow cytometry titrations.
Affinity determination: Measure the equilibrium dissociation constant (Kd) through Scatchard analysis or surface plasmon resonance. Properly conjugated antibodies show minimal affinity changes from their unconjugated counterparts .
Specificity verification: Confirm recognition of the intended epitope through Western blot analysis of REM1-expressing cells, expecting a single band at approximately 32.78 kDa .
Lot-to-lot consistency: Compare biotinylation degree, immunoreactivity, and specificity between production lots. Variation should be minimal to ensure experimental reproducibility.
Stability assessment: Monitor performance after storage under recommended conditions (-20°C or -80°C) over time. Activity should remain >90% for at least 6 months when properly aliquoted to avoid freeze-thaw cycles .
Aggregation analysis: Evaluate by size exclusion chromatography or dynamic light scattering. Minimal aggregation (<5%) ensures consistent performance across applications.
Rigorous application of these quality control metrics allows researchers to confidently interpret experimental results and ensure reproducibility across studies using biotin-conjugated REM1 antibodies.
Biotin-conjugated REM1 antibodies offer significant potential for pretargeted radioimmunotherapy (PRIT) applications, leveraging the biotin-streptavidin system to enhance therapeutic efficacy while reducing systemic toxicity. This advanced application follows a sophisticated multi-step methodology:
Initial targeting: The biotin-conjugated REM1 antibody is first administered to bind specifically to cells expressing REM1 protein, allowing time for tumor localization and blood clearance (typically 20 hours) .
Clearing agent administration: A biotin-N-acetyl-galactosamine clearing agent is then introduced to remove any circulating unbound antibody from the bloodstream. This clearing step improves the target-to-background ratio by complexing with and removing unbound antibody .
Radioisotope delivery: After clearing (typically 4 hours), radiolabeled biotin compounds such as 90Y-DOTA-biotin or 111In-DOTA-biotin are administered, which rapidly bind to the pre-localized biotin-conjugated antibody .
Enhanced tumor-to-normal tissue ratios: This approach has demonstrated superior tumor-to-normal organ ratios of radioactivity compared to conventional radioimmunotherapy. Research has shown tumor-to-blood ratios exceeding 65:1 after 48 hours using similar biotin-streptavidin pretargeting systems .
Therapeutic efficacy: With optimized protocols, more than 90% of tumor-bearing mice have been cured with minimal toxicity using 1200 μCi (44.4 MBq) 90Y-DOTA-biotin in similar pretargeting systems .
This methodology effectively separates the slow antibody distribution phase from the rapid radiation delivery phase, enabling higher radiation doses to be delivered to REM1-expressing tissues while sparing normal tissues from prolonged radiation exposure.
Multiplexed detection of REM1 alongside other biomarkers requires sophisticated methodological approaches that preserve the specificity of each detection system while enabling simultaneous visualization or quantification. The following strategies have been validated for multiplex applications with biotin-conjugated antibodies:
Sequential detection with enzyme inactivation: For immunohistochemistry applications, researchers can perform sequential rounds of biotin-based detection using different chromogens (DAB, AEC, etc.) with intermediate peroxidase inactivation steps (3% hydrogen peroxide, 10-15 minutes) between detection rounds.
Tyramide signal amplification (TSA) multiplexing: This approach uses HRP-catalyzed deposition of different fluorophore-labeled tyramides, allowing serial detection of multiple targets using the same biotin-streptavidin backbone. After each detection round, HRP activity is quenched before introducing the next biotin-conjugated antibody.
Spectrally distinct streptavidin conjugates: For flow cytometry or immunofluorescence, streptavidin conjugated to different fluorophores (e.g., streptavidin-allophycocyanin, streptavidin-phycoerythrin) can be used with biotin-conjugated REM1 antibodies and directly labeled antibodies against other targets.
Orthogonal labeling systems: Combining biotin-streptavidin detection for REM1 with other binding pairs (e.g., digoxigenin-anti-digoxigenin, DNP-anti-DNP) for other targets enables truly parallel multiplexing without cross-reactivity.
Mass cytometry (CyTOF): Streptavidin conjugated to rare earth metals can bind biotin-conjugated REM1 antibodies, enabling highly multiplexed detection alongside dozens of other markers without spectral overlap limitations.
These methodologies enable researchers to examine the relationship between REM1 expression and other biomarkers of interest, providing deeper insights into signaling pathway interactions and cellular phenotypes.
Biotin-conjugated REM1 antibodies offer powerful methodological approaches for investigating protein-protein interactions involving REM1, providing insights into its functional relationships within signaling networks. The following techniques leverage the biotin-conjugation to study these interactions:
Co-immunoprecipitation with streptavidin capture: Biotin-conjugated REM1 antibodies can be used to isolate intact protein complexes from cell lysates using streptavidin-conjugated magnetic beads. This approach maintains native protein interactions and can be coupled with mass spectrometry for unbiased identification of novel binding partners.
Proximity ligation assay (PLA): This technique can detect protein interactions with spatial resolution below 40 nm. By combining biotin-conjugated REM1 antibodies with antibodies against suspected interaction partners, followed by appropriate oligonucleotide-conjugated streptavidin and secondary antibodies, researchers can visualize specific interactions as distinct fluorescent spots.
BioID or APEX2 proximity labeling: Fusion of biotin ligase (BirA*) or APEX2 to REM1 enables biotinylation of proximal proteins, which can then be enriched using the same streptavidin systems employed with biotin-conjugated antibodies. This provides a complementary approach to validate interactions detected through antibody-based methods.
Fluorescence resonance energy transfer (FRET): Using biotin-conjugated REM1 antibodies with streptavidin-conjugated donor fluorophores alongside acceptor-labeled antibodies against potential interaction partners enables FRET measurements that can confirm direct protein-protein interactions within 10 nm.
Single-molecule pull-down: This technique combines biotin-streptavidin capture with single-molecule fluorescence imaging to analyze protein complexes at the individual molecule level, providing insights into interaction stoichiometry and heterogeneity.
These methodologically rigorous approaches enable researchers to map the interactome of REM1, providing critical insights into its role in cellular signaling networks and potential involvement in disease mechanisms.