CMIP Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Development of CMIP Antibody

CMIP (c-Maf-inducing protein) antibodies are immunological tools designed to detect and study the CMIP protein, which plays roles in T-cell signaling , cancer progression , and podocyte function . These antibodies are produced using recombinant or synthetic CMIP antigens, with various clones available for different applications. Key examples include:

Antibody CloneHost/TypeApplications (Tested)Reactivity
12851-1-AP (Proteintech)Rabbit/PolyclonalWB, IHC, IP, IF, ELISAHuman, Mouse, Rat
83381-1-RR (Proteintech)Rabbit/RecombinantWB, IHC, Flow Cytometry (Intra)Human
31407-1-AP (Proteintech)Rabbit/PolyclonalWB, IHCHuman

2.1. Western Blot (WB)

  • Detects endogenous CMIP at 86 kDa (full-length isoform) and 69–75 kDa (truncated isoform) .

  • Validated in cell lines including K-562, Raji, and A549 .

2.2. Immunohistochemistry (IHC)

  • Highlights CMIP in gastric cancer tissues, with overexpression linked to larger tumor size, lymph node metastasis, and poor survival .

  • Protocols recommend antigen retrieval using TE buffer (pH 9.0) or citrate buffer (pH 6.0) .

2.3. Functional Studies

  • Knockdown experiments: CMIP-siRNA reduced proliferation by 30.98% and increased apoptosis by 75.98% in gastric cancer cells (MKN-28) .

  • Mechanistic insights: CMIP inhibits Fyn/Lck kinases in T cells and promotes NF-κB inactivation in cancer .

3.2. Regulation of Signaling Pathways

PathwayCMIP FunctionExperimental Model
TCR ActivationInhibits Fyn/Lck kinases, blocks LAT recruitmentTransgenic mouse T cells
NF-κB SignalingBinds RelA, prevents nuclear translocationJurkat cells, podocytes
ApoptosisIncreases Bax/Bcl-2 ratio, activates caspase-3HEK cells, MN patient biopsies

4.1. Recommended Dilutions

Application12851-1-AP83381-1-RR31407-1-AP
Western Blot1:500–1:10001:5000–1:50,0001:2000–1:12,000
Immunohistochemistry1:50–1:5001:50–1:5001:50–1:500

Clinical Relevance and Future Directions

CMIP antibodies have enabled discoveries in:

  • Cancer biology: Targeting CMIP reduces metastasis in glioma and gastric cancer .

  • Nephrology: CMIP-WT1 interactions drive podocyte damage in nephrotic syndrome .

  • Therapeutic potential: siRNA-mediated CMIP knockdown is being explored for tumor suppression .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method and location. Please contact your local distributor for specific delivery times.
Synonyms
C-Maf-inducing protein antibody; c-Mip antibody; cmip antibody; CMIP_HUMAN antibody; KIAA1694 antibody; Tc-Mip antibody; TCMIP antibody; Truncated c-Maf-inducing protein antibody
Target Names
CMIP
Uniprot No.

Target Background

Function
CMIP plays a significant role in the T-cell signaling pathway. Isoform 2 may participate in the T-helper 2 (Th2) signaling pathway and appears to be the first proximal signaling protein linking T-cell receptor-mediated signal to the activation of the Th2-specific factor c-Maf.
Gene References Into Functions
  1. Genome-wide significant (P < 5 x 10(-8) ) interaction with MTNR1B and joint effects were detected for CMIP intronic rs17197883. PMID: 29691896
  2. Each copy of CMIP rs2925979_T allele was associated with a 29% higher risk of T2DM in females (p = 9.30 x 10(-4)), while it was not significantly associated with T2DM in males (p = 0.705). PMID: 29597287
  3. CMIP knockdown was observed to lead to the downregulation of MDM2 and mitogen activated protein kinase (MAPK) expression at the mRNA level, and CMIP was identified as a direct target of miR1013p. PMID: 28944848
  4. CMIP exhibits oncogenic properties and could potentially serve as a target for human glioma diagnosis and treatment. PMID: 28744466
  5. These findings suggest that CMIP haploinsufficiency is the likely cause of syndromic Autism spectrum disorder in our patients. PMID: 28504353
  6. Wilm's tumor protein (WT1) bound to 2 WT1 response elements, located at positions -290/-274 and -57/-41 relative to the transcription start site. PMID: 27650733
  7. Results indicate that c-mip and RelA define two distinct types of renal damage associated with VEGF-targeted therapies. PMID: 24067439
  8. c-mip plays an active role in the podocyte disorders of membranous nephropathy. PMID: 23302718
  9. These results suggest that alterations in NF-kappaB activity might stem from the up-regulation of c-mip and are likely to contribute to podocyte disorders in cases of idiopathic nephrotic syndrome. PMID: 22507836
  10. We present novel evidence that the language-disorder candidate gene CMIP is implicated in reading processes. PMID: 21457949
  11. Eight suggestive significant loci were detected with a series of genes expressed within the inner ear that underlie auditory function, such as: DCLK1, PTPRD, GRM8, CMIP. PMID: 21493956
  12. Results identify c-mip as a key component in the molecular pathogenesis of acquired podocyte diseases. PMID: 20484117
  13. Induction of c-mip in Hodgkin-Reed Sternberg cells and podocytes is associated with minimal change nephrotic syndrome in classical Hodgkin lymphoma. PMID: 20200355
  14. Results indicate that Tc-mip plays a critical role in the Th2 signaling pathway and represents the first proximal signaling protein that links TCR-mediated signal to the activation of c-maf Th2-specific factor. PMID: 12939343
  15. Filamin-A interacts with c-mip/Tc-mip in a novel T-cell signaling pathway. PMID: 15128042
  16. c-mip inhibits the degradation of I-kappaBalpha and impedes the dissociation of the NF-kappaB/I-kappaBalpha complexes. PMID: 19019440
  17. CMIP modulates phonological short-term memory in language impairment. PMID: 19646677

Show More

Hide All

Database Links

HGNC: 24319

OMIM: 610112

KEGG: hsa:80790

STRING: 9606.ENSP00000446100

UniGene: Hs.594095

Subcellular Location
Nucleus. Cytoplasm.
Tissue Specificity
Isoform 1 is expressed in peripheral blood mononuclear cells and kidney. Lower expression in brain and liver. Expression is down-regulated in activated cells. Isoform 2 is expressed in lymphocyte precursors, however, expression shuts down during maturatio

Q&A

What is CMIP and why is it significant in immunological research?

CMIP (C-Maf Inducing Protein) is an adapter protein that plays a crucial role in T-cell signaling pathways. Research has demonstrated that CMIP functions as a negative regulator of T cell signaling, inhibiting activation of the Src kinases Fyn and Lck after CD3/CD28 costimulation and subsequently blocking their localization to lipid rafts . CMIP is particularly significant because it drives T cells toward a naïve phenotype when selectively expressed in T cells, suggesting its important role in T cell development and function . Furthermore, CMIP has been implicated in several pathological conditions, including systemic lupus erythematosus and INS-like disease associated with cancers, making it a relevant target for immunological research . Understanding CMIP and having reliable antibodies against it is essential for investigating these disease mechanisms and potential therapeutic interventions.

What are the common applications for CMIP antibodies in research?

CMIP antibodies are utilized across multiple experimental applications in immunological and cell biology research. Based on the available antibody products, the most common applications include Western blotting (WB) for protein detection and quantification, immunohistochemistry (IHC) for visualizing CMIP distribution in tissue sections, immunoprecipitation (IP) for isolating CMIP and its binding partners, and immunofluorescence/immunocytochemistry (IF/ICC) for subcellular localization studies . Western blotting typically requires dilutions of 1:500 to 1:2000, while IHC applications generally use more concentrated antibody solutions (1:50 to 1:200) . These applications are critical for studying CMIP's role in T-cell signaling pathways, its interaction with binding partners like Fyn and the P85 subunit of PI3 kinase, and its cellular localization patterns under various conditions . CMIP antibodies have been successfully used to demonstrate that the protein can shuttle between the cytoplasm, plasma membrane (via its PH domain), and nuclear compartment (via its nuclear localization site) .

How do I select the appropriate CMIP antibody for my specific experimental needs?

Selecting the appropriate CMIP antibody requires careful consideration of several experimental factors. First, determine your target species (human, mouse, rat) and ensure the antibody has demonstrated reactivity against your species of interest . Next, consider the specific application (WB, IHC, IF, IP) and verify that the antibody has been validated for that particular method, as performance can vary significantly across applications .

If studying specific domains or isoforms of CMIP, select antibodies targeting relevant epitopes - for instance, antibodies targeting amino acids 157-274 versus those targeting the C-terminal region (AA 735-764) may yield different results depending on your research question . For research focusing on T-helper 2 (Th2) signaling pathways, consider antibodies specifically validated against isoform 2, which appears to link T-cell receptor-mediated signals to the activation of c-Maf Th2 specific factor .

Also consider the antibody format (conjugated vs. unconjugated) based on your detection method. Various conjugated forms (FITC, Biotin, HRP) are available for specific detection needs . Finally, review published literature using your antibody of choice to validate its performance in contexts similar to your experimental design.

What controls should I include when using CMIP antibodies in my experiments?

Proper experimental controls are essential when working with CMIP antibodies to ensure valid and reproducible results. For Western blotting and immunoprecipitation experiments, include positive controls such as cell lines known to express CMIP (like activated T cells) . Negative controls should include samples where CMIP expression is absent or significantly reduced, such as T cells from CMIP knockout or knockdown models . The search results mention a mouse model of selective and doxycycline-inducible CMIP ablation in T cells (CD2-rtTA/TetOn-Cre/CMIP loxP/loxP) that could provide excellent negative control material .

For immunohistochemistry applications, include isotype controls (using the same concentration of an irrelevant antibody of the same isotype) to assess non-specific binding. Additionally, considering that CMIP expression changes during T cell activation, comparing resting versus activated T cells can serve as physiological controls for antibody specificity . In all experiments, include technical replicates and validate findings using orthogonal methods when possible. For antibodies targeting specific CMIP epitopes, consider using peptide competition assays to confirm specificity, particularly when working with polyclonal antibodies that might recognize multiple epitopes .

How can I optimize immunoprecipitation protocols for CMIP to identify novel binding partners?

Optimizing immunoprecipitation (IP) protocols for CMIP requires careful consideration of several technical aspects to maximize sensitivity and specificity. Based on the mass spectrometry-based methods for antibody quality assessment, begin by selecting a high-quality CMIP antibody that has demonstrated IP efficiency . Consider antibodies classified as "IP gold standard," where the target antigen or known protein complex members are the most abundant proteins in the immunoprecipitate .

When designing your IP protocol, the cell lysis conditions are crucial since CMIP localizes to different cellular compartments. Use a lysis buffer that effectively solubilizes membrane-associated proteins while maintaining protein-protein interactions. Because CMIP interacts with lipid rafts and membrane components, consider using digitonin or CHAPS-based buffers that better preserve membrane protein associations compared to more stringent detergents like SDS or NP-40 .

To identify novel binding partners, implement a quantitative proteomic approach similar to the one described in search result . Compare normalized spectral abundance factors (NSAFs) from CMIP IP samples with control IPs. Cross-link the antibody to beads to reduce antibody contamination in the sample, which can mask lower-abundance interacting proteins during mass spectrometry analysis.

For validation of novel interactions, perform reciprocal IPs using antibodies against the potential binding partners. Additionally, consider proximity labeling approaches (BioID or APEX) as complementary methods to identify transient or weak CMIP interactions that might be lost during traditional IP procedures. When analyzing results, focus particularly on proteins involved in T-cell signaling pathways, as CMIP has established roles in regulating Src kinases Fyn and Lck, P85 subunit of PI3 kinase, RelA, filamin A, and DIP-1 .

What methodological approaches can resolve contradictory findings in CMIP subcellular localization studies?

Contradictory findings regarding CMIP subcellular localization can arise from various technical and biological factors. The research indicates that CMIP can shuttle between the cytoplasm, plasma membrane (via its PH domain), and nuclear compartment (via its nuclear localization site) . To resolve discrepancies in localization studies, implement a multi-faceted approach.

First, employ multiple, validated antibodies targeting different epitopes of CMIP. Different antibodies may have varying accessibility to epitopes depending on CMIP's conformation or interaction partners in different cellular compartments . Combine immunofluorescence with subcellular fractionation followed by Western blotting to provide complementary data on localization patterns.

Consider cellular activation state as a critical variable. As shown in the research, CMIP expression and localization are dynamically regulated during T cell activation, with endogenous CMIP levels rapidly falling upon CD3/CD28 stimulation while transgene-expressed CMIP may show different patterns . Therefore, carefully control and document the activation status of cells in your experiments.

For definitive studies, implement live-cell imaging using fluorescently tagged CMIP to track its movement between cellular compartments in real-time. This approach can reveal dynamic localization patterns that might be missed in fixed-cell studies. Additionally, create truncation or point mutants that disrupt specific domains (PH domain, nuclear localization signal) to demonstrate the functionality of these regions in controlling CMIP localization.

When interpreting results, consider cell type-specific differences. The research suggests that CMIP expression patterns and functions may vary across different cell types and pathological conditions . Finally, correlate localization patterns with functional readouts, such as effects on T cell signaling components or cytokine production, to establish the biological relevance of observed localization patterns.

How can I assess and troubleshoot CMIP antibody cross-reactivity with other protein family members?

Assessing and troubleshooting CMIP antibody cross-reactivity requires a systematic approach to ensure experimental specificity. Begin with in silico analysis by using tools like BLAST to identify proteins with sequence similarity to CMIP, paying particular attention to the specific epitope region targeted by your antibody . This computational approach can predict potential cross-reactive proteins.

For experimental validation, perform Western blotting using recombinant CMIP alongside structurally similar proteins. A critical experimental control involves using samples from CMIP knockout models, such as the doxycycline-inducible CMIP ablation T cell model mentioned in the research . In these samples, any remaining signal would indicate cross-reactivity with other proteins.

Consider implementing a mass spectrometry-based approach similar to that described in search result . This method quantifies the abundance of all proteins in immunoprecipitates and can identify non-specific binding. An antibody with high specificity would show CMIP as the most abundant protein in the immunoprecipitate, while cross-reactive antibodies would pull down multiple proteins at similar abundances.

If cross-reactivity is detected, several troubleshooting strategies can be employed. First, try increasing the stringency of your washing conditions in immunoprecipitation or Western blotting protocols. Second, perform peptide competition assays using the specific peptide used as the immunogen to confirm signal specificity . For polyclonal antibodies showing cross-reactivity, consider affinity purification against the specific CMIP epitope to enrich for antibodies with higher specificity.

Finally, validate your findings using orthogonal methods that do not rely on antibody recognition, such as mass spectrometry for protein identification or RNA-based methods (RT-qPCR, RNA-seq) to correlate protein detection with transcript levels. This multi-method approach can help distinguish true CMIP signals from cross-reactive artifacts.

What are the optimal conditions for detecting low-abundance CMIP in different tissue samples?

Detecting low-abundance CMIP in tissue samples requires optimized protocols to enhance sensitivity while maintaining specificity. Based on the search results and general principles of protein detection, implement these methodological approaches:

For immunohistochemistry (IHC), antigen retrieval is critical. The protocols mention high-pressure antigen retrieval in citrate buffer (pH 6.0) for paraffin-embedded tissues . Compare multiple antigen retrieval methods (heat-induced vs. enzymatic, different pH buffers) to determine optimal conditions for your specific tissue type. Signal amplification systems like HRP-conjugated streptavidin-biotin or tyramide signal amplification can significantly enhance detection sensitivity .

When performing Western blotting, optimize protein extraction by using buffers containing appropriate detergents (RIPA or NP-40 based) supplemented with protease inhibitors. Consider concentrating proteins from larger sample volumes using precipitation methods (TCA, acetone) or immunoprecipitation prior to Western blotting. Using higher antibody concentrations (1:200-1:500 range) may improve detection of low-abundance targets, though this increases the risk of non-specific binding .

For improved sensitivity in immunofluorescence, consider using high-sensitivity confocal microscopy with photomultiplier tubes or newer detection systems like Airyscan. Multi-layered detection systems, using primary antibody followed by biotinylated secondary and fluorophore-conjugated streptavidin, can amplify weak signals.

Regardless of the detection method, fresh or properly preserved samples are essential, as CMIP degradation may occur during extended storage. For all applications, blocking with at least 10% normal serum (matching the species of the secondary antibody) for 30 minutes at room temperature can reduce background and improve signal-to-noise ratio .

Finally, validate your detection protocol using positive control samples with known CMIP expression (such as certain T cell populations or pancreatic tissue) and negative controls like CMIP-deficient tissues or cells where CMIP has been knocked down.

How can I use CMIP antibodies to investigate its role in T-cell signaling pathways across different disease models?

Investigating CMIP's role in T-cell signaling across disease models requires integrating antibody-based detection with functional assays. The research indicates that CMIP is a negative regulator of T-cell signaling and inhibits activation of Src kinases Fyn and Lck after CD3/CD28 costimulation . To study this across disease models, develop a comprehensive experimental strategy.

Begin with baseline characterization of CMIP expression in your disease models using Western blotting and immunohistochemistry. The research shows that CMIP abundance increases in conditions like systemic lupus erythematosus and certain cancer-associated diseases . Quantify CMIP levels in patient samples compared to controls using RT-qPCR and Western blotting with appropriate CMIP antibodies .

For mechanistic studies, design immunoprecipitation experiments to assess CMIP's interaction with key T-cell signaling molecules (Fyn, Lck, PI3K, RelA) across disease states . Follow this with phosphorylation analysis of these proteins, as the research shows CMIP affects the phosphorylation status of Src kinases . For example, quantify the relative abundance of inactive forms (pY528Fyn, pY505Lck) versus active forms in CMIP-expressing versus control cells.

To assess functional consequences, isolate T cells from disease models and control subjects, then stimulate with anti-CD3/CD28 antibodies while monitoring downstream effects. Measure cytokine production (IL-2, IL-4, IFNγ, IL-10) as the research shows CMIP expression alters cytokine profiles . Additionally, examine T cell activation markers (CD44, CD62L) and proliferation rates to characterize the "naïve" T cell phenotype associated with CMIP expression .

For direct evidence of CMIP's role, implement gain-of-function approaches (CMIP overexpression) and loss-of-function strategies (CMIP knockdown/knockout) in your disease models. The doxycycline-inducible CMIP ablation system described in the research provides a useful model for controlled CMIP manipulation . Finally, use video microscopy analysis to visualize the recruitment of signaling molecules like LAT to the site of TCR engagement, as CMIP has been shown to block this process .

What biophysical techniques can complement antibody-based approaches to study CMIP structure-function relationships?

Biophysical techniques can provide crucial insights into CMIP structure-function relationships beyond what antibody-based approaches alone can reveal. While antibodies are excellent for detecting and localizing CMIP, integrating multiple biophysical methods provides a more comprehensive understanding of its molecular properties and interactions.

Circular dichroism (CD) spectroscopy can characterize CMIP's secondary structure components (α-helices, β-sheets) and monitor structural changes upon binding to partners or under different conditions. X-ray crystallography or cryo-electron microscopy would provide high-resolution structural information about CMIP, particularly focused on its key functional domains - the PH domain involved in membrane localization and the nuclear localization site .

Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) can quantitatively measure binding kinetics and affinities between purified CMIP and its interaction partners, including the Src kinases Fyn and Lck, the P85 subunit of PI3 kinase, RelA, and others identified in the research . These techniques allow determination of association/dissociation rates and binding constants under various conditions.

Hydrogen-deuterium exchange mass spectrometry (HDX-MS) can map CMIP conformational changes upon interaction with binding partners, revealing which regions undergo structural rearrangements during complex formation. This approach is particularly useful for identifying allosteric effects in protein interactions.

For studying CMIP's membrane interactions via its PH domain, techniques like lipid binding assays, liposome sedimentation assays, or microscale thermophoresis with lipid vesicles can characterize its lipid preferences and membrane association dynamics. Additionally, fluorescence resonance energy transfer (FRET) between fluorescently labeled CMIP and its binding partners can monitor interactions in live cells, complementing the antibody-based localization studies.

Finally, single-molecule techniques like total internal reflection fluorescence (TIRF) microscopy can track individual CMIP molecules in cellular contexts, revealing dynamic behaviors that might be masked in population-based studies. These approaches, used in conjunction with traditional antibody-based detection methods, provide a multi-dimensional view of CMIP structure and function.

How can I develop a quantitative assay to measure CMIP-mediated inhibition of T cell activation using CMIP antibodies?

Developing a quantitative assay to measure CMIP-mediated inhibition of T cell activation requires integrating antibody-based detection with functional readouts. Based on the research showing CMIP's role as a negative regulator of T cell signaling , design a comprehensive assay system with the following components:

First, establish a cell-based system with variable CMIP expression levels. This could include primary T cells from the CMIP transgenic mouse model described in the research , T cells from the doxycycline-inducible CMIP knockout model , or a cell line with inducible CMIP expression. Confirm and quantify CMIP expression levels in each condition using Western blotting with validated CMIP antibodies .

For activation measurements, stimulate T cells with anti-CD3/CD28 antibodies (1 μg/ml each) as described in the research . Design a time-course experiment (15, 30, 60 minutes) to capture early signaling events. Quantify the activation of proximal T cell signaling components using phospho-specific antibodies against key targets. Based on the research, focus particularly on:

  • The inactive forms of Src kinases (pY528Fyn, pY505Lck)

  • The active forms of signaling molecules (pY418 Src kinases, pY319Zap70)

Implement a flow cytometry-based assay to simultaneously measure multiple parameters in individual cells. This approach allows correlation between CMIP levels and activation markers on a per-cell basis. Use fluorescently labeled antibodies against both CMIP and phosphorylated signaling proteins. Additionally, include antibodies against T cell activation markers (CD69, CD25) for later time points.

For a more comprehensive analysis, measure downstream functional outcomes including:

  • Cytokine production (IL-2, IL-4, IFNγ, IL-10) using ELISA or intracellular cytokine staining, as the research shows CMIP alters cytokine profiles

  • T cell proliferation using CFSE dilution assays

  • Changes in T cell phenotype markers (CD44, CD62L) by flow cytometry

Develop a quantitative readout by calculating an "inhibition index" - the ratio of phosphorylated (active) to non-phosphorylated (inactive) signaling molecules as a function of CMIP expression level. Finally, validate your assay using known modulators of CMIP function or T cell signaling, and confirm key findings with orthogonal methods such as imaging to visualize the recruitment of signaling molecules to the immunological synapse.

What considerations are important when using CMIP antibodies for multiplex immunofluorescence with other T-cell signaling markers?

Multiplex immunofluorescence combining CMIP antibodies with other T-cell signaling markers requires careful technical considerations to ensure valid results. Based on the research and general principles of multiplex immunostaining, implement these methodological approaches:

Primary antibody selection is critical - choose CMIP antibodies and other T-cell signaling marker antibodies raised in different host species to avoid cross-reactivity between secondary antibodies . For example, if using a rabbit polyclonal CMIP antibody, select mouse monoclonal antibodies for other markers like Fyn, Lck, or LAT . When matching antibodies from the same species is unavoidable, consider directly conjugated primary antibodies or sequential staining with complete blocking steps between rounds.

Optimize the signal-to-noise ratio for each antibody individually before combining them. This includes determining the optimal antibody concentration, incubation time, and temperature for each target. The research indicates that CMIP antibodies are typically used at 1:50-1:200 dilutions for immunohistochemistry and 1:500-1:2000 for Western blotting , but each application may require specific optimization.

Spectral overlap between fluorophores is a major concern. Select fluorophores with minimal spectral overlap and include single-stained controls for spectral unmixing during analysis. For studying CMIP's co-localization with lipid rafts, as mentioned in the research , pair CMIP antibody detection with markers like cholera toxin B (for lipid rafts) using fluorophores with distinct emission spectra.

Sample preparation is critical - the research describes high-pressure antigen retrieval in citrate buffer (pH 6.0) for paraffin-embedded tissues . Ensure that antigen retrieval conditions are compatible with all target epitopes. Some epitopes may require different retrieval methods, necessitating sequential staining protocols.

For analyzing co-localization between CMIP and other signaling molecules (Fyn, Lck, LAT) , implement quantitative co-localization analysis using metrics like Pearson's correlation coefficient or Manders' overlap coefficient. This provides objective measurements of spatial relationships between molecules.

Finally, include appropriate controls for each experiment: positive controls (tissues/cells known to express each marker), negative controls (tissues/cells lacking expression), antibody isotype controls, and fluorescence-minus-one (FMO) controls to accurately set gating boundaries in flow cytometry or thresholds in image analysis.

How can I leverage CMIP antibodies in single-cell analysis techniques to study T cell heterogeneity?

Integrating CMIP antibodies into single-cell analysis techniques offers powerful approaches to dissect T cell heterogeneity at unprecedented resolution. Based on the research showing CMIP's selective expression in T cell subsets and its role in driving T cells toward a naïve phenotype , implement these methodological strategies:

For flow cytometry-based single-cell analysis, develop a multiparameter panel combining CMIP antibodies with markers of T cell subsets (CD4, CD8, CD44, CD62L) and activation states . Consider using spectral flow cytometry to accommodate more markers simultaneously. Optimize staining protocols with appropriate fixation and permeabilization methods, as CMIP localizes to multiple cellular compartments . Implement dimensionality reduction techniques like t-SNE or UMAP to visualize and identify cellular populations with distinct CMIP expression patterns.

For single-cell mass cytometry (CyTOF), conjugate CMIP antibodies to rare earth metals and combine with metal-labeled antibodies against T cell markers and signaling molecules (phosphorylated Fyn, Lck, Zap70) . CyTOF allows simultaneous measurement of over 40 parameters without spectral overlap concerns, enabling comprehensive characterization of CMIP-expressing cells.

Combining CMIP protein detection with transcriptomic analysis provides powerful insights into cellular heterogeneity. Implement CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) by using oligonucleotide-tagged CMIP antibodies alongside other tagged antibodies, allowing simultaneous measurement of surface/intracellular proteins and gene expression in single cells.

For spatial analysis of CMIP expression in tissues, employ multiplexed immunofluorescence imaging techniques like CODEX or Imaging Mass Cytometry. These methods can map CMIP expression relative to other markers with spatial context preserved, enabling identification of microanatomical niches where CMIP may play specific roles in T cell function.

Finally, develop computational pipelines for integrating these multimodal single-cell data. Apply trajectory inference algorithms to understand how CMIP expression changes during T cell activation and differentiation processes. This approach could reveal transition states and branch points where CMIP regulation is particularly crucial for T cell fate decisions.

What are the considerations for using CMIP antibodies in different species models given its evolutionary conservation?

Using CMIP antibodies across different species models requires careful consideration of evolutionary conservation, epitope preservation, and validation strategies. The search results indicate that some CMIP antibodies show cross-reactivity with human, mouse, and rat CMIP , suggesting evolutionary conservation of certain epitopes, but this requires systematic validation for each experimental system.

First, perform sequence alignment analysis of CMIP across species of interest to identify regions of high conservation that might serve as ideal epitope targets. Pay particular attention to the amino acid regions targeted by available antibodies (e.g., AA 157-274, AA 735-764) . Calculate percent identity and similarity scores for these epitope regions to predict potential cross-reactivity.

For each new species model, conduct thorough validation studies. Begin with Western blotting to confirm the antibody recognizes a protein of the expected molecular weight. Include positive controls (e.g., human samples) and negative controls (tissues/cells with CMIP knocked down) . For definitive validation, heterologous expression systems can be used - express the species-specific CMIP in a null background cell line and confirm antibody detection.

Be aware that even validated cross-reactive antibodies may have different optimal working conditions across species. Titrate antibody concentrations and optimize protocols for each species rather than assuming identical conditions. Additionally, consider that epitope accessibility may differ across species due to variations in post-translational modifications, protein folding, or interaction partners.

Finally, use orthogonal methods to confirm findings - pair antibody-based detection with nucleic acid detection methods (RT-qPCR, RNA-seq) to verify that protein detection correlates with species-specific transcript expression patterns. This multi-method approach strengthens confidence in cross-species applications of CMIP antibodies.

How can I apply mass spectrometry-based approaches to validate CMIP antibody specificity in complex biological samples?

Mass spectrometry-based approaches offer powerful tools for validating CMIP antibody specificity in complex biological samples. Based on the mass spectrometry methodology described in search result , implement a systematic validation strategy.

Start with immunoprecipitation (IP) using your CMIP antibody in the biological sample of interest. Include appropriate controls such as an isotype-matched irrelevant antibody and, if available, samples lacking CMIP expression . Process the immunoprecipitated proteins for mass spectrometry analysis following standardized protocols.

Implement the quantitative approach described in search result by comparing normalized spectral abundance factors (NSAFs) of all proteins in the immunoprecipitates. An antibody with high specificity will show CMIP as the most abundant protein in the IP sample, along with known interacting partners . Calculate enrichment factors by comparing the abundance of each protein in the CMIP antibody IP versus control IPs.

For comprehensive analysis, employ both data-dependent acquisition (DDA) for discovery and data-independent acquisition (DIA) or parallel reaction monitoring (PRM) for targeted quantification of CMIP and potential cross-reactive proteins. These targeted approaches offer increased sensitivity for detecting low-abundance proteins that might be missed in discovery-mode analysis.

For antibodies claiming to recognize specific post-translational modifications or isoforms, verify these claims through mass spectrometry. For instance, if studying the isoform 2 that plays a role in the T-helper 2 signaling pathway , confirm detection of isoform-specific peptides.

Finally, implement the "IP gold standard" classification described in search result , where antibodies are considered highly specific if the target antigen or a member of its known protein complex is the most abundant protein in the immunoprecipitate. This standardized approach allows objective assessment of antibody quality and specificity across different experimental conditions.

What strategies can improve reproducibility when using different lots of CMIP antibodies in longitudinal studies?

Ensuring reproducibility when using different antibody lots in longitudinal studies is critical for generating reliable scientific data. Based on general principles of antibody validation and the specific characteristics of CMIP antibodies, implement these methodological strategies:

Establish a comprehensive antibody validation protocol for each new lot. This should include Western blotting to confirm the expected molecular weight, immunohistochemistry or immunofluorescence to verify staining patterns, and quantitative assays to determine lot-to-lot variation in sensitivity . Document these validation results in a standardized format for future reference.

Create and maintain reference standards - these can be recombinant CMIP protein, cell lysates with known CMIP expression levels, or tissue sections with characterized CMIP distribution patterns . Test each new antibody lot against these standards under identical conditions. Calculate normalization factors between lots if absolute quantification is required.

When transitioning between lots, perform side-by-side comparisons using identical samples and protocols. Run parallel experiments with both the old and new antibody lots to generate overlapping data points that allow calibration between lots. This approach is particularly important for quantitative applications like Western blotting or flow cytometry.

Consider purchasing larger quantities of a single lot when planning longitudinal studies. Polyclonal antibodies are particularly susceptible to lot-to-lot variation due to their heterogeneous nature . Aliquot and store according to manufacturer recommendations to maintain stability throughout the study duration.

For critical applications, employ orthogonal detection methods alongside antibody-based approaches. Combine antibody detection with mass spectrometry-based protein quantification or nucleic acid measurements (RT-qPCR) to provide complementary data that can help normalize between antibody lots.

Implement standardized reporting of antibody information in all experimental records and publications. Include catalog numbers, lot numbers, validation data, and detailed experimental conditions . This documentation facilitates troubleshooting if reproducibility issues arise and enables more accurate replication by other researchers.

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 2025 TheBiotek. All Rights Reserved.