SOCS7 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

SOCS7 recombinant monoclonal antibodies are laboratory-produced immunoglobulins engineered using recombinant DNA technology. These antibodies are generated by cloning genes encoding the variable regions of heavy and light chains into expression vectors, which are transfected into host cells (e.g., HEK-293) for large-scale production . They specifically target SOCS7, a protein involved in:

  • Insulin signaling: Mediates ubiquitination and degradation of IRS1 to regulate glucose homeostasis .

  • Cytokine inhibition: Suppresses STAT3/5 activation by sequestering them in the cytoplasm, modulating prolactin, growth hormone, and leptin pathways .

  • Ubiquitination: Acts as a substrate-recognition component in E3 ubiquitin ligase complexes to degrade proteins like DAB1, influencing neuronal development .

Key Features:

  • Immunogen: Synthetic peptides or recombinant fragments derived from human SOCS7 (e.g., amino acids 1–100 or 1–200) .

  • Purification: Affinity chromatography (Protein A/G) .

  • Storage: Typically stable at -20°C in glycerol-containing buffers to prevent freeze-thaw damage .

A. Functional Studies

  • Insulin Resistance: SOCS7 knockdown enhances insulin sensitivity by stabilizing IRS1, making it a potential therapeutic target for metabolic disorders .

  • Neuronal Development: SOCS7 regulates reelin signaling by degrading phosphorylated DAB1, critical for cortical neuron positioning .

B. Diagnostic and Therapeutic Potential

  • Cancer Research: SOCS7 is implicated in glioblastoma and breast adenocarcinoma, with overexpression observed in tumor cell lines (e.g., U-87 MG, MCF7) .

  • Immunohistochemistry (IHC): Validated in human brain and testis tissues, showing cytoplasmic and membrane localization .

Validation Data

AssayPerformanceSource
Western Blot (WB)Detects 63 kDa band in human cell lysates (HeLa, MCF7, Jurkat) .
IHC-PStaining observed in paraffin-embedded human brain and testis at 1:50 dilution .
Flow Cytometry (FC)Specific binding to HepG2 cells confirmed via FITC-conjugated secondary antibodies .

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The SOCS7 recombinant monoclonal antibody is produced through in vitro cloning. This process involves integrating genes encoding both the heavy and light chains of the SOCS7 antibody into expression vectors. These vectors are then introduced into host cells to enable the expression of the recombinant antibody within a cell culture environment. Following expression, the SOCS7 recombinant monoclonal antibody is purified from the supernatant of transfected host cell lines using an affinity chromatography-based purification method. This antibody exhibits high specificity in binding to the human SOCS7 protein, making it suitable for diverse applications including ELISA, IHC, and FC.

SOCS7 is a negative regulator of cytokine and growth factor signaling pathways, primarily by participating in the JAK-STAT pathway. Its function is to prevent excessive immune responses, regulate cell growth and differentiation, and maintain homeostasis. Dysregulation of SOCS7 can contribute to various diseases and conditions.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery time may vary depending on the chosen purchase method or location. Please consult your local distributors for specific delivery timelines.
Synonyms
Suppressor of cytokine signaling 7 (SOCS-7) (Nck, Ash and phospholipase C gamma-binding protein) (Nck-associated protein 4) (NAP-4), SOCS7, NAP4 SOCS6
Target Names
Uniprot No.

Target Background

Function

SOCS7 regulates signaling cascades potentially through protein ubiquitination and/or sequestration. It plays a role in insulin signaling and glucose homeostasis by promoting the ubiquitination of IRS1, leading to its subsequent proteasomal degradation. SOCS7 also inhibits prolactin, growth hormone, and leptin signaling by preventing STAT3 and STAT5 activation. This inhibition is achieved by sequestering these proteins in the cytoplasm and reducing their binding to DNA. SOCS7 may serve as a substrate recognition component of a SCF-like E3 ubiquitin-protein ligase complex, which mediates the ubiquitination and subsequent proteasomal degradation of target proteins.

Gene References Into Functions
  1. This study demonstrated elevated protein levels of SOCS-4 and SOCS-7 in Alzheimer's disease brains. PMID: 25286386
  2. The observed effect of SOCS7 knockdown could be attributed to a specific anti-PLCg-1 role. PMID: 25162020
  3. Genetic variants in the SOCS7 gene do not appear to significantly influence variation in glucose homeostasis traits or the risk of T2DM in the Old Order Amish population. PMID: 23767996
  4. Treatment with Insulin Like Growth Factor I and SOCS7 loss have synergistically resulted in enhanced growth and migration of MCF7 cells and increased migration of MDA-MB-231 cells. PMID: 24046004
  5. This report presents, for the first time, associations between common variants in the SOCS7 gene and obesity, central obesity, insulin resistance, and disorders of lipid metabolism in nondiabetic men from Argentina. PMID: 22397880
  6. SOCS7 functions as an oncogene, revealing a novel mechanism of carcinogenesis in bladder cancer cells. PMID: 23392170
  7. LNCaP-S17 cells exhibit resistance to exogenous IL-6-induced neuroendocrine differentiation due to increased levels of CIS/SOCS7, which block the activation of JAK2-STAT3 pathways. PMID: 22213096
  8. Higher mRNA expression levels of SOCS1, 3, 4, and 7 are significantly associated with earlier tumor stage and improved clinical outcome in human breast cancer. PMID: 20433750
  9. Findings suggest that PPAR-gamma is involved in the regulation of SOCS-7 expression by hepatitis C virus core protein genotype 3a. PMID: 20357037
  10. Our data indicate that SOCS-7 interacts with vinexin and the actin cytoskeleton. PMID: 15242778
  11. Results suggest that SOCS7 is a potent regulator of prolactin, leptin, and growth hormone signaling. Its mode of action involves inhibiting cytokine-inducible STAT3 and 5-mediated signal transduction. PMID: 15677474
  12. Data suggest that SOCS7 is a significant regulator of glucose homeostasis and insulin signaling. PMID: 16127460

Show More

Hide All

Database Links

HGNC: 29846

OMIM: 608788

KEGG: hsa:30837

STRING: 9606.ENSP00000464034

UniGene: Hs.514132

Subcellular Location
Cytoplasm. Cell membrane; Peripheral membrane protein; Cytoplasmic side. Nucleus. Note=Mostly cytoplasmic, but shuttles between the cytoplasm and the nucleus. Rapidly relocalizes to the nucleus after UV irradiation. Cytoplasmic location depends upon SEPT7 presence.
Tissue Specificity
Expressed in brain and leukocytes. Also in fetal lung fibroblasts and fetal brain.

Q&A

What is a SOCS7 Recombinant Monoclonal Antibody and how does it differ from traditional monoclonal antibodies?

SOCS7 Recombinant Monoclonal Antibodies are laboratory-engineered antibodies created using recombinant DNA technology rather than traditional hybridoma methods. They are specifically designed to target the SOCS7 protein, which plays a critical role in signal transduction pathways as a negative regulator of cytokine signaling. Unlike traditional monoclonal antibodies produced in animals, recombinant monoclonal antibodies are generated from defined DNA sequences and can be produced in various expression systems such as HEK293 cells, which provides several advantages. The primary benefit is complete control over the antibody sequence, allowing researchers to customize specificity, species reactivity, and other properties for specific experimental needs.

The generation of recombinant monoclonal antibodies begins with identifying the primary amino acid sequence of the antibody through techniques like transcriptome shotgun sequencing or isolating antigen-specific antibody-secreting cells. This DNA sequence can then be cloned into expression vectors and transfected into mammalian cells for production. For SOCS7 antibodies, this typically involves a rabbit IgG backbone with carefully selected binding regions that recognize specific epitopes within the human SOCS7 protein structure. The recombinant nature ensures batch-to-batch consistency and eliminates concerns about hybridoma stability or drift over time.

The purification process typically employs affinity chromatography methods to isolate the antibody from expression media, resulting in high purity and specificity. Products like the CSB-RA594821A0HU SOCS7 Recombinant Monoclonal Antibody (clone 23E4) are formulated with appropriate buffers like phosphate-buffered saline with glycerol and preservatives to maintain stability and function. This recombinant approach addresses many of the reproducibility and standardization issues that plague traditional animal-derived antibodies, making them valuable tools for consistent research applications .

What are the validated applications for SOCS7 Recombinant Monoclonal Antibodies in research settings?

SOCS7 Recombinant Monoclonal Antibodies have been extensively validated for several critical research applications, with immunohistochemistry (IHC), flow cytometry (FC), and enzyme-linked immunosorbent assay (ELISA) being the primary validated methods. For immunohistochemistry applications, these antibodies have demonstrated effective staining in paraffin-embedded human tissues, particularly in testis tissue as shown with the CSB-RA594821A0HU antibody when used at an optimized dilution of 1:50. The IHC application typically involves antigen retrieval under high pressure in citrate buffer (pH 6.0), followed by blocking with normal goat serum, and detection using polymer-based secondary antibody systems with DAB visualization .

In flow cytometry applications, SOCS7 antibodies enable researchers to quantitatively analyze SOCS7 expression at the single-cell level, providing information about protein expression heterogeneity within cell populations. Optimal dilutions for flow cytometry typically range from 1:50 to 1:200, with protocols involving cell fixation in formaldehyde, permeabilization with detergents like Triton X-100, and blocking with normal serum prior to antibody incubation. The ability to perform multiparameter analysis makes flow cytometry particularly valuable for studying SOCS7 in relation to other signaling molecules within the same cells.

For ELISA applications, these antibodies allow for quantitative detection of SOCS7 protein in biological samples, though specific protocols and sensitivities may vary between different antibody clones. Beyond these primary applications, researchers may adapt SOCS7 antibodies for Western blotting, immunoprecipitation, or ChIP assays, though additional validation may be necessary for these extended applications. The versatility of these antibodies across multiple techniques makes them valuable tools for comprehensive investigation of SOCS7 biology in human cells and tissues .

What optimization strategies are recommended for immunohistochemistry with SOCS7 Recombinant Monoclonal Antibodies?

Successful immunohistochemistry with SOCS7 Recombinant Monoclonal Antibodies requires careful optimization of several key parameters to achieve specific staining with minimal background. Based on validated protocols, antigen retrieval represents a critical step that significantly impacts staining quality. For SOCS7 detection in paraffin-embedded tissues, high-pressure heat-induced epitope retrieval using citrate buffer (pH 6.0) has proven effective in unmasking the epitopes that may be cross-linked during formalin fixation. The duration and temperature of this step should be carefully titrated, as insufficient retrieval may result in false negatives while excessive treatment can distort tissue morphology and increase non-specific binding.

Blocking protocols require particular attention when working with SOCS7 antibodies. A 30-minute room temperature incubation with 10% normal goat serum has been validated to effectively reduce background staining by blocking non-specific binding sites. The concentration and incubation conditions for the primary antibody are crucial determinants of staining specificity and intensity. For the CSB-RA594821A0HU SOCS7 antibody, dilutions between 1:50 and 1:200, with overnight incubation at 4°C in 1% BSA solution, provide optimal results in many tissue types .

Detection systems should be carefully selected to provide adequate sensitivity without amplifying background signals. HRP-labeled polymer-based detection systems have proven effective for SOCS7 IHC, with standardized development using 0.43% DAB. When optimizing new tissue types or experimental conditions, researchers should implement appropriate positive controls (tissues known to express SOCS7) and negative controls (omission of primary antibody) to validate the specificity of staining patterns. Finally, counterstaining protocols and mounting media selection should be optimized to provide adequate contrast for visualization of positive signals while preserving long-term stability of the stained sections.

How should researchers properly store and handle SOCS7 Recombinant Monoclonal Antibodies to maintain activity?

Proper storage and handling of SOCS7 Recombinant Monoclonal Antibodies are essential for maintaining their activity and ensuring consistent experimental results over time. Upon receipt, these antibodies should be stored at -20°C or -80°C in their original containers to prevent degradation of the protein structure and loss of binding specificity. Repeated freeze-thaw cycles can significantly damage antibody structure and function, so it is recommended to aliquot the stock solution into smaller volumes based on typical experimental usage to minimize the number of freeze-thaw events. Each aliquot should be clearly labeled with the antibody name, catalog number, lot number, concentration, and date of preparation.

The buffer composition plays a crucial role in maintaining antibody stability during storage. The CSB-RA594821A0HU SOCS7 antibody, for example, is formulated in phosphate-buffered saline (pH 7.4) containing 150mM NaCl, 0.02% sodium azide, and 50% glycerol. The high glycerol content prevents freezing at -20°C, allowing for easier handling while maintaining stability, while the sodium azide serves as a preservative to prevent microbial contamination . When preparing working dilutions from the stock solution, use high-quality, sterile buffers free from contaminants that could interfere with antibody binding or cause degradation.

For daily experimental use, antibody working solutions should be kept on ice or at refrigerated temperatures (2-8°C) and used within the recommended time frame, typically within 12-24 hours of dilution. When handling the antibody, use appropriate laboratory practices including the use of clean, nuclease-free tubes and pipette tips to prevent contamination. Additionally, researchers should maintain detailed records of antibody usage, including freeze-thaw cycles, to facilitate troubleshooting if inconsistent results are observed. Following these storage and handling guidelines will help ensure the longevity and consistent performance of SOCS7 recombinant monoclonal antibodies across experimental applications.

How can researchers validate the specificity of SOCS7 Recombinant Monoclonal Antibodies for their experimental systems?

Validating the specificity of SOCS7 Recombinant Monoclonal Antibodies is a critical prerequisite for generating reliable and reproducible research data. A comprehensive validation approach should employ multiple complementary methods to confirm that the antibody genuinely detects the intended target without cross-reactivity to related proteins. One powerful validation strategy involves using positive and negative control samples with known SOCS7 expression profiles. For instance, researchers can utilize SOCS7-overexpressing cell lines (such as stably transfected HEK293T cells) as positive controls and compare their staining patterns with parental untransfected cells or SOCS7 knockout cells generated through CRISPR-Cas9 technology as negative controls.

Western blot analysis serves as a critical validation tool, allowing researchers to confirm that the antibody detects a protein of the expected molecular weight (approximately 64 kDa for human SOCS7). The presence of a single distinct band at the appropriate molecular weight strongly supports antibody specificity, while multiple bands may indicate cross-reactivity or protein degradation requiring further investigation. For immunohistochemistry applications, researchers should perform peptide competition assays, wherein pre-incubation of the antibody with the immunizing peptide should abolish or significantly reduce specific staining if the antibody is truly binding to its intended epitope.

RNA interference approaches provide another powerful validation method, where siRNA or shRNA-mediated knockdown of SOCS7 expression should result in corresponding reduction of signal intensity with a specific antibody. Similarly, correlation between protein detection and mRNA expression data (from RT-PCR or RNA-seq) across different tissues or experimental conditions provides additional confirmation of specificity. Implementing this multi-faceted validation approach enables researchers to confidently attribute their experimental observations to genuine SOCS7 biology rather than artifacts stemming from antibody cross-reactivity or non-specific binding .

What methodological considerations are important for flow cytometry analysis using SOCS7 Recombinant Monoclonal Antibodies?

Flow cytometry analysis using SOCS7 Recombinant Monoclonal Antibodies requires careful attention to several methodological factors to obtain accurate and interpretable data. Cell fixation and permeabilization protocols are particularly critical since SOCS7 is an intracellular protein involved in signaling pathways. Based on validated protocols, optimal results have been achieved using 4% formaldehyde fixation followed by permeabilization with 0.2% Triton X-100, which provides adequate access to intracellular epitopes while preserving cellular morphology and antibody binding sites. The duration and temperature of fixation (typically 10-15 minutes at room temperature) should be carefully controlled to avoid overfixation, which can mask epitopes and reduce antibody binding.

Appropriate blocking procedures are essential to minimize non-specific antibody binding and reduce background fluorescence. A 30-minute blocking step using 10% normal goat serum has been demonstrated to effectively reduce background in flow cytometry applications with SOCS7 antibodies. Primary antibody concentration and incubation conditions significantly impact staining quality, with optimal results typically achieved using a 1:50 dilution and incubation for 45 minutes at 4°C. Selection of appropriate secondary antibodies is equally important, with FITC-conjugated goat anti-rabbit IgG at 1:200 dilution providing effective detection for rabbit-derived SOCS7 antibodies .

Proper controls are indispensable for accurate flow cytometry analysis. Isotype controls (such as rabbit IgG at equivalent concentration to the primary antibody) are essential to establish baseline fluorescence and identify non-specific binding. Additionally, positive controls (cells known to express SOCS7) and negative controls (cells with low or no SOCS7 expression) help validate staining patterns and facilitate accurate gating strategies. When analyzing data, researchers should implement compensation protocols to correct for spectral overlap when performing multicolor flow cytometry. Finally, consistent instrument settings and calibration are crucial for comparing SOCS7 expression across different experimental conditions or time points.

What are the approaches to reduce potential immunogenicity when using recombinant monoclonal antibodies in research models?

Reducing potential immunogenicity of recombinant monoclonal antibodies, including those targeting SOCS7, is critical for research applications involving animal models or long-term in vitro studies. Several strategic approaches can be implemented to minimize unwanted immune responses that might confound experimental results. Humanization of antibody sequences represents a primary strategy for reducing immunogenicity in human cell systems or humanized animal models. This process involves replacing non-human framework regions with human sequences while retaining only the critical complementarity-determining regions (CDRs) responsible for antigen binding, significantly reducing the potential for anti-antibody responses while preserving target specificity.

Species matching represents a third critical consideration, particularly for in vivo applications. When designing experiments in animal models, researchers should select antibodies with constant regions matching the host species (e.g., murine constant regions for mouse models) to minimize recognition as foreign by the host immune system. Additionally, fragment-based approaches offer advantages for reducing immunogenicity in certain applications. Using smaller antibody fragments such as Fab fragments, single-chain variable fragments (scFv), or nanobodies can significantly reduce immunogenicity by eliminating the constant regions that often contain immunogenic epitopes while retaining target binding capability. Finally, careful purification to remove aggregates, endotoxins, and other contaminants that might enhance immune responses is essential for any recombinant antibody preparation used in research .

How can researchers optimize the use of SOCS7 Recombinant Monoclonal Antibodies for specific tissue types?

Optimizing SOCS7 Recombinant Monoclonal Antibodies for specific tissue types requires systematic adaptation of protocols to account for the unique properties of each tissue. Tissue-specific fixation protocols represent the first critical parameter for optimization. While 10% neutral buffered formalin is commonly used, the optimal fixation duration may vary significantly between tissues due to differences in density and penetration rates. For instance, delicate tissues like lymphoid organs may require shorter fixation times (4-12 hours) compared to dense tissues like brain or liver (24-48 hours). Overfixation can mask SOCS7 epitopes through excessive protein cross-linking, while insufficient fixation may result in poor tissue morphology and antigen preservation.

Antigen retrieval methods must be optimized for each tissue type to effectively unmask epitopes while preserving tissue architecture. While citrate buffer (pH 6.0) with high-pressure heat treatment has been validated for testis tissue , other tissues may respond better to different retrieval buffers such as EDTA (pH 9.0) or Tris-EDTA (pH 8.0). The optimal retrieval duration and temperature should be determined empirically for each tissue type through systematic comparison of different conditions. Tissue-specific blocking strategies are equally important, as different tissues contain varying levels of endogenous peroxidases, biotin, and proteins that can contribute to background staining.

Antibody dilution and incubation conditions often require tissue-specific optimization. While a 1:50 dilution has proven effective for testis tissue immunohistochemistry , other tissues with different SOCS7 expression levels or accessibility may require adjusted concentrations. Researchers should perform systematic dilution series (e.g., 1:25, 1:50, 1:100, 1:200) for each new tissue type to identify the optimal concentration that maximizes specific signal while minimizing background. Finally, counterstaining protocols should be adapted to provide optimal contrast for each tissue type, with lighter counterstaining often beneficial for tissues with expected low SOCS7 expression to avoid obscuring positive signals.

What approaches can be used to adapt SOCS7 Recombinant Monoclonal Antibodies for super-resolution microscopy applications?

Adapting SOCS7 Recombinant Monoclonal Antibodies for super-resolution microscopy applications requires specialized approaches to overcome the resolution limitations imposed by conventional antibody labeling. The size of traditional full-length antibodies (approximately 10-15 nm) can significantly compromise the achievable resolution in techniques like PALM/STORM or STED microscopy, where spatial resolutions of 20-30 nm are possible. To address this limitation, researchers can employ antibody fragments to reduce the distance between the fluorophore and the target epitope. Single-chain variable fragments (scFv), which contain only the variable regions of the heavy and light chains linked by a flexible peptide, provide much smaller probes (approximately 2-3 nm) that can substantially improve the precision of target localization in super-resolution microscopy.

Direct fluorophore conjugation strategies are particularly valuable for super-resolution applications with SOCS7 antibodies. By directly coupling fluorophores to the antibody rather than using secondary detection methods, researchers eliminate the additional spatial displacement introduced by secondary antibodies, which can add another 10-15 nm to the distance between the fluorophore and the target. Site-specific labeling approaches, where fluorophores are conjugated at defined positions away from the antigen-binding site, can further improve the precision of target localization. For optimal results in super-resolution imaging, specialized fluorophores with appropriate photophysical properties should be selected, such as those with high quantum yield, photostability, and suitable blinking characteristics for techniques like STORM .

The density of labeling is another critical consideration for super-resolution microscopy with SOCS7 antibodies. Under-labeling can result in incomplete visualization of structures, while over-labeling may lead to excessive background or overlapping signals that compromise resolution. Researchers should systematically optimize antibody concentration and incubation conditions to achieve an appropriate labeling density for their specific application. Additionally, sample preparation techniques such as expansion microscopy can be combined with SOCS7 antibody labeling to physically expand the specimen, effectively increasing the distance between fluorophores and improving the apparent resolution without requiring specialized microscopy equipment .

How should researchers quantify and interpret immunohistochemistry data from SOCS7 Recombinant Monoclonal Antibody staining?

Quantification and interpretation of immunohistochemistry data from SOCS7 Recombinant Monoclonal Antibody staining requires structured analytical approaches to generate reliable and reproducible results. Systematic scoring systems should be established prior to analysis to minimize subjective bias. H-score methods, which combine staining intensity (0-3+) and percentage of positive cells to generate scores ranging from 0-300, provide a comprehensive quantitative assessment of SOCS7 expression. Alternatively, researchers may employ quickscore systems that separately evaluate the proportion of positively stained cells and staining intensity, or simpler approaches that categorize expression as negative, weak, moderate, or strong based on predefined criteria specific to SOCS7 staining patterns.

Digital image analysis offers advantages for objective quantification of SOCS7 expression in immunohistochemistry. Using specialized software, researchers can set specific thresholds for positive staining based on color, intensity, and morphological parameters. This approach enables precise measurement of parameters such as percentage of positive cells, staining intensity, and subcellular localization patterns. For valid comparisons across different experimental conditions or tissue samples, standardized image acquisition parameters must be maintained, including consistent exposure times, white balance, and magnification. All images should be acquired using calibrated microscopy systems with proper quality control.

Interpretation of SOCS7 staining patterns requires consideration of both subcellular localization and context-specific expression. SOCS7 may exhibit different localization patterns depending on activation state and cell type, ranging from predominantly cytoplasmic to nuclear distribution. These localization patterns may have functional significance and should be reported alongside quantitative intensity data. Additionally, researchers should compare SOCS7 expression across different cell types within the same tissue section, as differential expression between adjacent cells may provide insights into tissue-specific regulatory mechanisms. Finally, all quantitative data should be subjected to appropriate statistical analysis, with clear reporting of the methods used for quantification, the number of fields or cells analyzed, and measures of variability.

What challenges exist in analyzing flow cytometry data for SOCS7 expression and how can they be addressed?

Flow cytometry analysis of SOCS7 expression presents several unique challenges that researchers must address to obtain reliable data. The intracellular nature of SOCS7 necessitates permeabilization procedures that can introduce variability and artifacts if not carefully controlled. Different permeabilization reagents may yield varying degrees of epitope accessibility while preserving or altering cellular autofluorescence characteristics. To address this challenge, researchers should systematically compare different permeabilization protocols (e.g., saponin, Triton X-100, methanol) to identify the optimal approach for their specific cell type and experimental conditions, always maintaining consistent protocols across experimental groups for valid comparisons.

Multiparameter analysis involving SOCS7 requires careful panel design to avoid spectral overlap between fluorophores. When SOCS7 staining is combined with markers for cell identity, activation state, or other signaling molecules, compensation between fluorophores becomes critical. Proper compensation controls, including single-stained samples for each fluorophore, must be included in each experiment. Additionally, fluorescence-minus-one (FMO) controls, where all fluorophores except SOCS7 are included, are essential for accurate gating and distinguishing positive SOCS7 signal from background in multicolor experiments .

Quantitative interpretation of SOCS7 flow cytometry data presents another challenge, particularly when comparing expression levels across different experimental conditions. Simple percentage of positive cells may be insufficient when SOCS7 expression changes in magnitude rather than in an all-or-none fashion. Median fluorescence intensity (MFI) provides a more nuanced measure of expression level, but requires standardization across experiments using calibration beads. For the most rigorous analysis, researchers should calculate molecules of equivalent soluble fluorochrome (MESF) values using calibration standards, which provides an absolute quantification that can be compared between experiments and instruments. Finally, appropriate statistical methods must be applied to flow cytometry data, with consideration of whether parametric or non-parametric tests are most appropriate based on the distribution of the data.

How can researchers integrate data from multiple applications of SOCS7 Recombinant Monoclonal Antibodies for comprehensive analysis?

Integrating data from multiple applications of SOCS7 Recombinant Monoclonal Antibodies enables researchers to develop a comprehensive understanding of SOCS7 biology that transcends the limitations of any single experimental approach. Correlation analysis between different detection methods represents a powerful strategy for data integration. For instance, researchers can perform quantitative comparisons between SOCS7 protein levels detected by immunohistochemistry, flow cytometry, and Western blotting across the same experimental conditions. This approach not only validates findings through multiple independent methods but also provides complementary information about expression levels, subcellular localization, and protein integrity that no single method can deliver alone.

Multi-scale integration of SOCS7 data from tissue to cellular to subcellular levels provides another valuable perspective. Immunohistochemistry data reveals tissue-level patterns and architectural context of SOCS7 expression, while flow cytometry enables quantitative single-cell analysis across large populations, and super-resolution microscopy uncovers nanoscale details of subcellular localization and protein complexes. By systematically collecting and analyzing data across these different scales, researchers can map relationships between tissue-level phenomena and underlying cellular and molecular mechanisms involving SOCS7.

Computational approaches can significantly enhance the integration of multimodal SOCS7 data. Machine learning algorithms can identify patterns and correlations across diverse datasets that might not be apparent through conventional analysis. For example, researchers can apply clustering algorithms to identify cell subpopulations with distinct SOCS7 expression patterns across different experimental conditions or diseased states. Network analysis approaches can integrate SOCS7 expression data with information about interacting proteins, signaling pathways, and functional outcomes to place SOCS7 within its broader biological context. Finally, temporal integration through time-course experiments using multiple detection methods can reveal dynamic aspects of SOCS7 regulation and function that would not be apparent from static measurements.

What are the key considerations for experimental design when studying SOCS7 function using recombinant monoclonal antibodies?

Designing robust experiments to study SOCS7 function using recombinant monoclonal antibodies requires careful consideration of several critical factors to ensure valid and interpretable results. Appropriate control selection stands as a fundamental element of experimental design that directly impacts data quality and interpretability. Experimental designs should incorporate positive controls (tissues or cells known to express high levels of SOCS7), negative controls (tissues or cells with minimal SOCS7 expression), technical controls (omission of primary antibody, isotype controls), and biological controls relevant to the specific research question (e.g., treatment vs. vehicle, wild-type vs. knockout, normal vs. diseased tissue). These comprehensive controls enable researchers to distinguish specific SOCS7 signals from technical artifacts and establish the biological significance of observed differences.

Time course considerations are particularly important when studying SOCS7, as this signaling regulator exhibits dynamic expression patterns in response to various stimuli. Experimental designs should include multiple time points to capture both rapid, early changes and delayed, sustained alterations in SOCS7 expression or localization. The temporal resolution required may vary based on the specific signaling pathway being investigated, with some cytokine responses inducing changes within minutes to hours, while developmental or chronic disease processes may require extended time courses spanning days to weeks. Signal amplification strategies represent another key consideration, as SOCS7 may be expressed at low levels in some cell types or under certain conditions.

Quantification strategies should be determined during experimental design rather than post hoc. Researchers should establish clear criteria for what constitutes positive SOCS7 staining, how intensity will be measured, and what statistical approaches will be used to analyze the data. When designing experiments involving manipulation of SOCS7 expression (overexpression, knockdown, or knockout), researchers should consider the timing of intervention relative to readout, potential compensatory mechanisms that may emerge, and appropriate controls for the method of genetic manipulation. Finally, replication strategies should be explicitly planned, including both technical replicates (repeated measurements within the same biological sample) and biological replicates (independent samples subjected to the same experimental conditions).

How can researchers effectively combine SOCS7 Recombinant Monoclonal Antibodies with other antibodies for co-localization studies?

Combining SOCS7 Recombinant Monoclonal Antibodies with other antibodies for co-localization studies requires strategic planning to avoid technical pitfalls while maximizing the biological insights gained from simultaneous detection of multiple proteins. Antibody compatibility assessment is the first critical consideration, as antibodies used in combination must be compatible in terms of species origin, isotype, and required experimental conditions. For example, if using a rabbit-derived SOCS7 antibody like CSB-RA594821A0HU, researchers should select companion antibodies raised in different species (e.g., mouse, rat, or goat) to enable selective detection with species-specific secondary antibodies without cross-reactivity. When same-species antibodies must be used, sequential staining protocols with intermediate blocking steps or directly conjugated primary antibodies offer potential solutions .

Optimization of detection systems is essential for achieving clear distinction between signals from different antibodies. For immunofluorescence co-localization studies, fluorophore selection should account for spectral properties to minimize bleed-through between channels. Fluorophores with well-separated excitation and emission spectra (e.g., FITC/Alexa 488 for one antibody and Cy3/Alexa 555 for another) are preferable, and appropriate single-stained controls must be included to establish correct exposure settings and verify the absence of spectral overlap. For chromogenic detection in brightfield microscopy, distinct chromogens (e.g., DAB for SOCS7 and Fast Red for a co-marker) with different colorimetric properties should be selected, along with optimization of the sequence of detection to prevent masking of epitopes.

Analysis of co-localization data requires rigorous quantitative approaches rather than subjective visual assessment. Researchers should employ specialized co-localization software to calculate parameters such as Pearson's correlation coefficient, Manders' overlap coefficient, or object-based co-localization metrics that provide objective measures of spatial relationship between SOCS7 and other proteins of interest. When interpreting co-localization data, the resolution limits of the imaging system must be considered, as conventional light microscopy cannot resolve structures closer than approximately 200 nm. For definitive evidence of molecular-scale co-localization, super-resolution techniques, proximity ligation assays, or complementary biochemical approaches such as co-immunoprecipitation should be employed to supplement standard co-localization imaging.

What approaches are recommended for investigating SOCS7 expression in rare cell populations?

Investigating SOCS7 expression in rare cell populations presents unique challenges that require specialized approaches to obtain reliable data from limited samples. Flow cytometry with cell enrichment stands as a powerful primary approach for analyzing SOCS7 in rare populations, allowing quantitative single-cell analysis even when target cells comprise less than 0.1% of the total population. Prior enrichment through techniques such as fluorescence-activated cell sorting (FACS), magnetic-activated cell sorting (MACS), or density gradient separation can significantly increase the yield of rare target cells. Using CSB-RA594821A0HU SOCS7 antibody in combination with lineage-specific surface markers enables identification and characterization of rare SOCS7-expressing subpopulations, with optimized staining protocols employing increased antibody concentrations and extended incubation times to enhance detection sensitivity .

Laser capture microdissection (LCM) offers a valuable approach for studying SOCS7 in rare cells within tissue contexts, allowing precise isolation of specific cell types identified by morphology or preliminary staining. Following LCM, isolated cells can be analyzed for SOCS7 expression using highly sensitive methods such as quantitative PCR, targeted proteomics, or even single-cell sequencing to correlate SOCS7 protein expression with transcriptomic profiles. This approach is particularly valuable for rare cell types that cannot be effectively isolated by flow cytometry due to lack of specific surface markers or sensitivity to enzymatic dissociation procedures.

How can researchers establish correlations between SOCS7 protein levels and functional outcomes in signaling pathways?

Establishing meaningful correlations between SOCS7 protein levels and functional outcomes in signaling pathways requires integrated experimental approaches that connect molecular detection with functional readouts. Phosphorylation state analysis of signaling components represents a primary approach, as SOCS7 functions as a negative regulator in several signaling cascades. By simultaneously measuring SOCS7 expression and the phosphorylation status of downstream effectors such as JAK/STAT pathway components using phospho-specific antibodies, researchers can directly correlate SOCS7 levels with pathway activation states. This can be accomplished through multiplexed flow cytometry, which enables single-cell correlation analysis, or through parallel Western blotting of samples under various conditions to establish quantitative relationships between SOCS7 expression and signaling pathway activity.

Genetic manipulation approaches provide powerful tools for establishing causal relationships between SOCS7 and signaling outcomes. By creating cellular models with controlled SOCS7 expression through techniques such as CRISPR-Cas9 knockout, siRNA knockdown, or inducible overexpression systems, researchers can observe how precisely manipulated SOCS7 levels impact downstream signaling events. These approaches are particularly valuable when combined with dose-response studies, where varying levels of pathway stimulation (e.g., different cytokine concentrations) are tested against different SOCS7 expression levels to map out the quantitative relationship between the regulator and the signaling response.

Time-resolved correlation studies are essential for understanding the dynamic relationship between SOCS7 and signaling pathways. Given that negative regulators often function within feedback loops, the temporal relationship between SOCS7 expression and pathway activity may involve complex dynamics that cannot be captured at a single time point. Experimental designs should include synchronized time course measurements of both SOCS7 levels and functional readouts following pathway stimulation. Mathematical modeling approaches can then be applied to time-resolved data to infer causal relationships and feedback mechanisms. Finally, single-cell correlation analysis using techniques such as imaging flow cytometry or multiplexed immunofluorescence enables researchers to account for cell-to-cell heterogeneity in both SOCS7 expression and signaling responses, revealing correlations that might be obscured in population-averaged measurements.

What emerging technologies are enhancing the capabilities of recombinant monoclonal antibodies for research applications?

Emerging technologies are dramatically expanding the capabilities of recombinant monoclonal antibodies, including those targeting SOCS7, for advanced research applications. Single-cell antibody discovery platforms represent a revolutionary approach that enables rapid isolation and cloning of antibodies with desired specificities. These technologies, such as transcriptionally active PCR (TAP), allow direct generation of recombinant antibodies from individual antigen-specific antibody secreting cells (ASCs). This approach significantly accelerates the process of obtaining functional monoclonal antibodies while simultaneously enabling comprehensive analysis of variable region repertoires in combination with functional assays to identify antibodies with optimal characteristics for specific research applications .

AI-driven antibody engineering is transforming how researchers design and optimize recombinant antibodies. Machine learning algorithms trained on vast antibody sequence datasets can predict structure-function relationships and guide rational design of antibodies with enhanced specificity, affinity, or reduced immunogenicity. These computational approaches can identify optimal mutation sites to improve binding properties or stability without compromising function, accelerating the development of next-generation recombinant antibodies for challenging targets like SOCS7. When combined with high-throughput experimental validation, these AI-driven approaches significantly reduce the time and resources required to develop highly optimized recombinant antibodies.

Multiplexed antibody technologies are enabling simultaneous detection of numerous targets within the same sample. Techniques such as cyclic immunofluorescence (CycIF), multiplexed ion beam imaging (MIBI), and co-detection by indexing (CODEX) allow for the detection of dozens to hundreds of proteins in a single tissue section using recombinant antibodies with defined specificities. These methods rely on sequential staining-imaging-bleaching cycles, metal-conjugated antibodies detected by mass spectrometry, or DNA-barcoded antibodies, respectively. Such approaches will enable researchers to place SOCS7 expression and localization within a complex cellular context, revealing its relationship to numerous other proteins simultaneously and providing unprecedented insights into its role in signaling networks and disease processes .

How are genetically encoded antibody fragments revolutionizing intracellular studies of signaling molecules like SOCS7?

Genetically encoded antibody fragments are transforming intracellular studies of signaling molecules like SOCS7 by enabling dynamic visualization and manipulation of endogenous proteins in living cells. Unlike conventional antibodies that cannot access intracellular targets in living cells, genetically encoded fragments such as single-chain variable fragments (scFvs), nanobodies, and designed ankyrin repeat proteins (DARPins) can be expressed directly within cells to bind their targets in the native cellular environment. These tools enable real-time monitoring of SOCS7 dynamics, including changes in expression, localization, and protein-protein interactions in response to signaling events, providing insights that are impossible to obtain with traditional fixed-cell approaches or biochemical methods.

Fluorescent protein fusions with antibody fragments create versatile intracellular sensors for monitoring SOCS7 in living systems. By genetically fusing fragments like scFvs to fluorescent proteins, researchers can generate conformational sensors that report on SOCS7 activation states or interaction events through changes in fluorescence resonance energy transfer (FRET) or fluorescence complementation. Similarly, "frankenbodies" - engineered scFvs optimized for intracellular stability and function - can be developed by grafting the hypervariable domains from SOCS7-specific antibodies onto validated scFv scaffolds. These constructs can be expressed in cells to track dynamic changes in endogenous SOCS7 localization and concentration over time in response to various stimuli .

Intracellular antibody fragments also enable functional perturbation of SOCS7 with unprecedented spatial and temporal precision. When expressed as intrabodies, these fragments can be designed to block specific protein-protein interactions or functional domains of SOCS7, creating highly specific loss-of-function conditions that target particular aspects of SOCS7 function rather than eliminating the entire protein. By combining these tools with inducible expression systems or optogenetic control elements, researchers can achieve precise temporal control over when and where SOCS7 function is perturbed. Furthermore, antibody-based protein degradation systems such as AbTACs (antibody-based targeted protein degradation) can be developed using SOCS7-specific antibody fragments to induce rapid, selective degradation of endogenous SOCS7 protein, providing a powerful complement to genetic knockout approaches for studying SOCS7 function in diverse cellular contexts.

What are the future prospects for implementing digital PCR technologies in SOCS7 recombinant antibody production and validation?

Digital PCR (dPCR) technologies offer transformative potential for enhancing both production and validation of SOCS7 recombinant monoclonal antibodies through unprecedented precision in nucleic acid quantification and analysis. In antibody sequence recovery and cloning, dPCR provides superior accuracy for quantifying and analyzing rare antibody transcripts from hybridoma cells or isolated B cells. The partitioning of reactions into thousands or millions of individual chambers in dPCR enables absolute quantification of antibody gene templates without standard curves, facilitating more precise amplification of variable regions from limited starting material. This capability is particularly valuable when recovering sequences from rare or degraded samples, potentially increasing the success rate of obtaining complete and accurate sequences for SOCS7-specific antibodies from single antibody-secreting cells.

For recombinant expression system optimization, dPCR enables precise monitoring of antibody gene expression levels in production platforms such as HEK293 cells. By accurately quantifying transgene copy number and expression levels, researchers can systematically optimize transfection conditions, select high-producing clones, and monitor production stability with unprecedented precision. This capability supports the development of more efficient and consistent manufacturing processes for recombinant SOCS7 antibodies, contributing to enhanced batch-to-batch reproducibility and reduced production costs through optimization of expression conditions.

In the validation domain, dPCR will enable more sensitive and precise correlation between SOCS7 protein levels (detected by the antibody) and SOCS7 mRNA expression. By providing absolute quantification of SOCS7 transcript levels across different tissues or experimental conditions, dPCR generates gold-standard reference data against which antibody-based protein detection can be validated. Additionally, the high sensitivity of dPCR allows detection of rare transcript variants or mutations in the SOCS7 gene that might affect epitope recognition, providing critical information for interpreting antibody binding patterns in genetically diverse samples. As single-cell technologies continue to evolve, integration of dPCR with microfluidic platforms will enable paired analysis of SOCS7 antibody binding and gene expression at the single-cell level, creating powerful new validation paradigms that link antibody performance directly to the molecular characteristics of individual cells.

How might synthetic biology approaches expand the functionality of recombinant antibodies for studying SOCS7 signaling networks?

Synthetic biology approaches are poised to dramatically expand the functionality of recombinant antibodies for studying SOCS7 signaling networks through the creation of engineered molecular tools with novel capabilities beyond conventional antibody functions. Multi-domain fusion proteins represent one promising direction, where SOCS7-specific binding domains from recombinant antibodies are genetically fused with additional functional domains to create chimeric proteins with enhanced or novel capabilities. For example, researchers could generate SOCS7-targeting constructs fused with enzymatic domains (such as HRP, luciferase, or BirA biotin ligase) for proximity labeling applications, enabling identification of proteins that interact with SOCS7 in specific cellular compartments or under different signaling conditions. Similarly, fusions with DNA-binding domains could create synthetic transcription factors that activate reporter gene expression in response to SOCS7 detection, translating protein expression into easily measured outputs.

Split-protein complementation systems based on antibody recognition offer powerful tools for studying SOCS7 protein-protein interactions in living cells. By splitting reporter proteins (such as luciferase or fluorescent proteins) and fusing each fragment to SOCS7-specific antibody fragments and putative interaction partners respectively, researchers can develop assays where reporter activity is reconstituted only when SOCS7 interacts with its binding partner. Such systems enable quantitative, real-time monitoring of dynamic protein interactions in living cells and can be adapted to high-throughput screening formats to identify modulators of SOCS7 interaction networks.

Synthetic circuit design incorporates SOCS7-specific antibody fragments into engineered cellular signaling pathways to create sophisticated biosensors or cellular devices with programmed responses to SOCS7 activity. By connecting antibody-based SOCS7 detection to synthetic transcriptional or post-translational regulatory elements, researchers can develop cells that respond to changes in SOCS7 expression or activation with precisely engineered outputs, such as expression of reporter genes, secretion of detectable markers, or initiation of therapeutic responses. These synthetic biology approaches transcend traditional analytical applications of antibodies, transforming SOCS7-specific recombinant antibodies from passive detection tools into active components of engineered biological systems that both sense and respond to changes in SOCS7 biology.

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.