LCMT1 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Introduction to LCMT1 and Biotin Conjugation

LCMT1 (leucine carboxyl methyltransferase 1) is an enzyme critical for post-translational methylation of PP2A phosphatase subunits, regulating their activity in cell signaling, survival, and differentiation . Biotin-conjugated LCMT1 antibodies are engineered to link anti-LCMT1 antibodies with biotin, enabling detection via streptavidin-based systems in assays like ELISA, Western blot (WB), and immunohistochemistry (IHC). This conjugation enhances sensitivity and amplification capabilities, particularly for low-abundance targets .

Product Characteristics of LCMT1 Biotin-Conjugated Antibodies

Below is a comparative analysis of commercially available LCMT1 biotin-conjugated antibodies:

ParameterProteintech (30222-1-AP) OriGene (Clone OTI4A5)
Host/IsotypeRabbit polyclonal IgGMouse monoclonal
ApplicationsWB, IHC, ELISAWB, IHC, ELISA
ReactivityHuman, MouseHuman, Mouse
Molecular Weight38–41 kDa (observed)Not specified
SupplierProteintech (China)OriGene Technologies (China)
Dilution RecommendationsWB: 1:500–1:3000; IHC: 1:50–1:500 Not explicitly stated

Role in Hematopoiesis and Cancer Research

  • Hematopoietic Defects: Global LCMT1 knockout in mice causes severe fetal hematopoiesis defects, including reduced PP2A B subunit stability and core heterodimer levels (~30% reduction) . Biotin-conjugated LCMT1 antibodies could track these changes in HSCs (hematopoietic stem cells) and lineage-committed progenitors.

  • Cancer Cell Survival: LCMT1 overexpression in breast (MCF7, MDA-MB-231) and pancreatic cancer cells correlates with resistance to LCMT1 inhibitors (e.g., Compound 2), as shown by clonogenic assays . Biotin-conjugated antibodies aid in quantifying LCMT1 levels to assess therapeutic responses.

Technical Advantages in Assays

Assay TypeApplication ExampleAdvantage
Western BlotDetecting LCMT1 in A549, HEK-293, or MCF7 lysates High sensitivity for low-abundance LCMT1; compatible with streptavidin-HRP
ImmunohistochemistryStaining mouse uterus tissue or tumor xenografts Requires antigen retrieval (TE buffer pH 9.0 or citrate buffer pH 6.0)
ELISAQuantifying LCMT1 in serum or lysatesAmplified signal via biotin-streptavidin interaction; high specificity

Conjugation Process

Biotin conjugation typically involves:

  1. Lyophilization: Pre-prepared biotin mix (e.g., LYNX Rapid Plus Biotin Kits) .

  2. Activation: Proprietary reagents enable antibody-biotin coupling at near-neutral pH.

  3. No Desalting Required: High efficiency (~100% antibody recovery) and rapid labeling (minutes) .

Optimization Tips

  • Antigen Retrieval: For IHC, use TE buffer (pH 9.0) or citrate buffer (pH 6.0) for optimal LCMT1 staining .

  • Titration: Dilute antibodies between 1:50–1:500 (IHC) or 1:500–1:3000 (WB) to minimize background .

Supplier and Availability

SupplierProduct CodeReactivityKey Features
Proteintech30222-1-APHuman, MouseAffinity-purified, liquid format
OriGene TechnologiesOTI4A5 (Biotin)Human, MouseMonoclonal, suitable for WB/IHC

Pricing: Proteintech offers next-day delivery for $40 , while OriGene lists products without explicit pricing .

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. The delivery time may vary depending on the purchase method or location. For specific delivery time, please contact your local distributor.
Synonyms
Leucine carboxyl methyltransferase 1 (EC 2.1.1.233) (Protein-leucine O-methyltransferase) ([Phosphatase 2A protein]-leucine-carboxy methyltransferase 1), LCMT1, LCMT
Target Names
LCMT1
Uniprot No.

Target Background

Function
This antibody targets the carboxyl group of the C-terminal leucine residue of protein phosphatase 2A catalytic subunits, modifying it to form alpha-leucine ester residues.
Gene References Into Functions
  1. Downregulation of LCMT1 may lead to alterations in the membrane localization of PP2A and Tau, potentially resulting in PP2A and Tau dysfunction in Alzheimer's disease. PMID: 23943618
  2. Crystal structure analysis of human LCMT-1, both in isolation and in complex with PP2A stabilized by a cofactor mimic, revealed that the active site of LCMT-1 specifically recognizes the carboxyl terminus of PP2A. PMID: 21292165
  3. The X-ray crystal structure of human LCMT1 protein in complex with the cofactor S-adenosylmethionine (AdoMet) has been determined to a resolution of 2 A. PMID: 21206058
  4. LCMT-1 plays a significant role in normal progression through mitosis and cell survival, and it is essential for embryonic development. PMID: 17724024
Database Links

HGNC: 17557

OMIM: 610286

KEGG: hsa:51451

STRING: 9606.ENSP00000382021

UniGene: Hs.337730

Protein Families
Methyltransferase superfamily, LCMT family

Q&A

What is LCMT1 and why is it significant in research?

LCMT1 (Leucine Carboxyl Methyltransferase 1) is an enzyme that methylates protein phosphatase catalytic subunits at their C-terminal leucine residues, primarily affecting PP2A (Protein Phosphatase 2A) and related phosphatases such as PP4 and PP6 . This methylation is crucial for regulating phosphatase complex assembly and function. LCMT1 is particularly significant in research because global knockout studies have demonstrated its essential role in embryonic development, with loss causing severe defects in fetal hematopoiesis and typically resulting in embryonic lethality by day 16.5 . Understanding LCMT1 function provides insights into fundamental cellular regulatory mechanisms involving protein phosphatases.

What experimental applications are most suitable for biotin-conjugated LCMT1 antibodies?

Biotin-conjugated LCMT1 antibodies are particularly valuable for enhanced detection sensitivity in techniques requiring signal amplification . They excel in immunohistochemistry applications where tissue samples may have low LCMT1 expression levels. The biotin-avidin/streptavidin detection system enhances signal without increasing background, making these conjugates ideal for ELISA, immunoprecipitation coupled with mass spectrometry, and immunohistochemical detection in tissue sections . Their application is especially valuable when studying LCMT1's role in methylation-dependent formation of phosphatase complexes, as documented in studies of PP2A BAC heterotrimers.

How should I validate the specificity of my LCMT1 antibody?

Proper validation of LCMT1 antibody specificity should involve multiple complementary approaches. First, perform Western blot analysis using both LCMT1 wild-type and knockout cell lysates (such as MEFs) to confirm antibody specificity by the absence of signal in knockout samples . Second, conduct pre-adsorption experiments using recombinant LCMT1 protein (aa 157-357) to block specific binding . Third, include positive controls consisting of tissues or cell lines known to express LCMT1. Fourth, perform immunoprecipitation followed by mass spectrometry to confirm that the antibody pulls down authentic LCMT1. Fifth, cross-validate results using additional LCMT1 antibodies raised against different epitopes .

What are the key considerations for optimizing immunofluorescence protocols with biotin-conjugated LCMT1 antibodies?

When optimizing immunofluorescence protocols with biotin-conjugated LCMT1 antibodies, several factors require careful consideration. Begin with appropriate fixation - paraformaldehyde (4%) is typically effective for preserving LCMT1 epitopes while maintaining cellular architecture . Block endogenous biotin using avidin/biotin blocking kits to prevent non-specific binding, particularly important in tissues with high endogenous biotin content (liver, kidney). Optimize antibody concentration through titration experiments (typically starting at 1-5 μg/mL) and determine optimal incubation time (4°C overnight often yields better results than shorter room temperature incubations) . Include a nuclear counterstain to provide cellular context for LCMT1 localization. Finally, validate subcellular localization patterns by comparison with published data on LCMT1 distribution.

How can I accurately assess LCMT1-dependent methylation of PP2A and other phosphatases using biotin-conjugated antibodies?

Accurately assessing LCMT1-dependent methylation of phosphatases requires a sophisticated multi-step approach. First, establish a baseline by conducting immunoprecipitation of the target phosphatases (PP2Ac, PP4c, PP6c) followed by Western blotting with methylation-sensitive antibodies from control samples . For enhanced detection sensitivity, utilize biotin-conjugated secondary antibodies in combination with streptavidin-HRP. To specifically attribute methylation to LCMT1 activity, compare phosphatase methylation levels between wild-type and LCMT1-deficient samples (using knockout or knockdown models) . For quantitative assessment, implement base treatment controls: divide cell lysates into two portions, treating one with base (which removes carboxyl methylation) followed by neutralization, and treating the other with pre-neutralized base solution to preserve methylation . This controlled comparison provides a measure of methylation status. Finally, confirm findings through methylation-dependent complex formation assays, as LCMT1-dependent methylation significantly impacts phosphatase complex assembly.

What strategies should be employed to investigate the relationship between LCMT1 expression and PP2A BAC holoenzyme assembly in disease models?

Investigation of LCMT1 expression and PP2A BAC holoenzyme assembly in disease models requires a comprehensive experimental strategy. Begin with quantitative assessment of LCMT1 expression levels across relevant disease and control tissues using biotin-conjugated LCMT1 antibodies for immunohistochemistry with signal amplification . Next, perform co-immunoprecipitation experiments using antibodies against B subunits (such as 2G9 monoclonal that detects Bα) followed by Western blotting for PP2Ac to quantify holoenzyme assembly . Blue Native-PAGE analysis provides superior resolution of intact phosphatase complexes - apply this technique to compare the distribution of PP2A complexes between diseased and healthy states . For functional correlation, measure phosphatase activity using phosphosubstrate assays specific to B-subunit directed activity. Finally, implement genetic rescue experiments by restoring LCMT1 expression in disease models to determine if PP2A complex formation and function can be normalized. This approach has proven effective in studies showing that LCMT1 loss reduces PP2A BAC heterotrimers by approximately 75% in knockout MEFs compared to wild-type cells .

How can apparent discrepancies between in vitro and in vivo studies regarding LCMT1-dependent methylation of phosphatases be resolved?

Resolving discrepancies between in vitro and in vivo studies of LCMT1-dependent methylation requires systematic investigation of contextual factors. First, recognize that in vitro studies have shown PP2A BAC heterotrimers can form without methylation, while in vivo studies demonstrate that LCMT1 knockout reduces these complexes by approximately 92% . This fundamental discrepancy likely arises from the cellular environment's complexity. To resolve this, conduct comparative studies using purified components in vitro with gradually increasing complexity by adding potential competing factors. Investigate the role of cellular factors like α4, which has been shown to interact with unmethylated PP2Ac and may represent competing binding interactions absent in simplified in vitro systems . Implement advanced structural biology approaches (cryo-EM, X-ray crystallography) to visualize the molecular interfaces between methylated/unmethylated phosphatases and regulatory subunits. Finally, develop cellular systems with controllable expression of methylation-site PP2A mutants to determine the precise contribution of this modification to complex formation in intact cells versus purified systems.

What are the methodological considerations for using biotin-conjugated LCMT1 antibodies to study temporal dynamics of LCMT1 expression during hematopoiesis?

Studying temporal dynamics of LCMT1 expression during hematopoiesis with biotin-conjugated antibodies requires sophisticated methodological considerations. First, establish a developmental timeline with precise staging of hematopoietic tissues from embryonic day 10.5 through 16.5, when LCMT1 knockout causes severe defects in fetal hematopoiesis . Implement multiparameter flow cytometry combining biotin-conjugated LCMT1 antibodies (detected with streptavidin-fluorophore conjugates) with hematopoietic stem cell (HSC) and progenitor markers (KLS: c-Kit+, Lineage-, Sca-1+) to track LCMT1 expression across differentiation stages . For tissue section analysis, employ multiplexed immunohistochemistry using tyramide signal amplification to simultaneously visualize LCMT1 and lineage markers. Crucially, validate antibody specificity within each developmental stage using LCMT1 heterozygous tissues as controls for expression levels . Finally, complement protein-level studies with transcriptional analysis through single-cell RNA sequencing to correlate LCMT1 expression with differentiation trajectories. This comprehensive approach captures the complex relationship between LCMT1 and hematopoietic development, building on findings that LCMT1 knockout reduces KLS cells and causes multilineage defects in fetal liver lineage analysis .

How can background signal be minimized when using biotin-conjugated LCMT1 antibodies in tissues with high endogenous biotin?

Minimizing background signal when using biotin-conjugated antibodies in biotin-rich tissues requires implementing a systematic multi-step strategy. First, incorporate a comprehensive avidin-biotin blocking step before primary antibody application - apply avidin solution (15-30 minutes), wash thoroughly, then apply biotin solution (15-30 minutes) to block all endogenous biotin and biotin-binding sites . Second, optimize fixation protocols to preserve antigen accessibility while minimizing tissue autofluorescence - brief fixation (10-15 minutes) with fresh 4% paraformaldehyde often provides optimal results. Third, include additional blocking steps with both serum (5-10% from the secondary antibody host species) and commercial biotin blocking solutions. Fourth, reduce primary antibody concentration to the minimum effective level determined through careful titration experiments . Fifth, implement stringent washing steps (6-8 washes of 5-10 minutes each) with PBS containing 0.1-0.3% Triton X-100 to remove unbound antibody. Finally, consider alternative detection strategies such as polymer-based detection systems if background persists despite these measures.

What strategies can address inconsistent results when comparing LCMT1 detection across different experimental systems?

Addressing inconsistent LCMT1 detection across experimental systems requires systematic troubleshooting focused on protocol standardization and biological variability. First, standardize sample preparation by implementing consistent lysis conditions optimized for LCMT1 preservation - phosphatase inhibitors are particularly important as PP2A activity may influence LCMT1 stability . Second, account for LCMT1 expression variability by normalizing to appropriate housekeeping proteins and including positive control samples with known LCMT1 expression in each experiment. Third, validate antibody performance in each system by confirming specificity through LCMT1 knockdown/knockout controls specific to each experimental context . Fourth, optimize antibody concentrations independently for each detection system - antibody requirements often differ significantly between Western blot, IHC, and flow cytometry applications . Fifth, implement quantitative analysis methods using digital imaging and standardized exposure settings to objectively compare signal intensity across experiments. Finally, consider the influence of post-translational modifications on LCMT1 epitope accessibility, as these may vary across experimental systems and affect antibody recognition.

How should researchers interpret changes in LCMT1 detection when studying PP2A complex assembly in disease models?

Interpreting changes in LCMT1 detection when studying PP2A complex assembly requires careful consideration of multiple factors affecting this regulatory relationship. First, establish whether observed changes reflect alterations in LCMT1 expression, activity, or localization by combining techniques - Western blot for total expression, activity assays for functional assessment, and subcellular fractionation for localization . Second, determine the consequential impact on PP2A methylation using methylation-specific antibodies and base treatment controls as established methodologically . Third, quantify changes in PP2A subunit levels (A, B, C) as LCMT1 loss reduces not only methylation but also total levels of these proteins in knockout models . Fourth, assess PP2A complex formation through blue native PAGE and co-immunoprecipitation experiments to determine if observed changes in LCMT1 correlate with altered complex assembly . Fifth, evaluate functional consequences through phosphatase activity assays specific to different PP2A complexes. Finally, implement genetic rescue experiments by modulating LCMT1 levels to establish causality between LCMT1 changes and observed PP2A complex alterations. This comprehensive interpretation framework helps distinguish between correlation and causation in disease models.

What are the key considerations for developing a multiplexed immunofluorescence protocol incorporating biotin-conjugated LCMT1 antibodies?

Developing effective multiplexed immunofluorescence protocols with biotin-conjugated LCMT1 antibodies requires addressing several technical challenges. First, carefully plan antibody combinations to avoid host species overlap - ideally select primary antibodies from different host species for each target (e.g., rabbit anti-LCMT1 paired with mouse antibodies against other targets) . Second, implement sequential detection for biotin-conjugated antibodies - complete the streptavidin-based detection of LCMT1 before introducing other biotin-containing reagents. Third, optimize signal separation through appropriate fluorophore selection - choose fluorophores with minimal spectral overlap and include single-color controls for spectral unmixing algorithms. Fourth, establish an optimized antigen retrieval protocol that works effectively for all target antigens, typically beginning with citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) heat-induced epitope retrieval. Fifth, validate staining patterns independently before combining antibodies, as multiplexing may introduce unexpected cross-reactivity or interference. Finally, implement rigorous controls including absorption controls with recombinant proteins and secondary-only controls to ensure specificity in the multiplexed setting .

How can researchers determine if observed hematopoietic defects are directly attributable to LCMT1 loss versus secondary effects on PP2A activity?

Determining whether hematopoietic defects result directly from LCMT1 loss versus secondary effects on PP2A activity requires a multifaceted investigative approach. First, conduct detailed temporal analysis of LCMT1 and PP2A activity throughout hematopoietic development to establish whether LCMT1 loss precedes changes in PP2A activity or complex formation . Second, implement rescue experiments using wild-type LCMT1 versus catalytically inactive LCMT1 mutants to determine if enzymatic activity is essential for normal hematopoiesis. Third, perform comparative phenotypic analysis between LCMT1 knockout models and models with targeted PP2A subunit modifications - similar phenotypes would suggest PP2A-mediated effects . Fourth, conduct phosphoproteomic analysis to identify altered phosphorylation patterns in hematopoietic stem cells following LCMT1 loss, comparing these to known PP2A substrate profiles. Fifth, utilize conditional tissue-specific knockout models to distinguish between intrinsic hematopoietic effects and secondary effects from supporting cell types. Research has shown that LCMT1 knockout causes severe defects in fetal hematopoiesis with reduction in KLS cells and multilineage defects in fetal liver lineage analysis, suggesting roles in HSC generation, self-renewal, and/or survival .

What are the implications of observed α4-PP2Ac complex formation in LCMT1 knockout models for experimental design?

The increased formation of α4-PP2Ac complexes in LCMT1 knockout models has significant implications for experimental design. First, recognize that standard co-immunoprecipitation approaches may fail to capture the full spectrum of PP2A complex redistribution following LCMT1 loss - blue native PAGE analysis reveals a high molecular weight PP2Ac-containing complex lacking B subunits but containing α4 in LCMT1 knockout models . Second, incorporate α4 analysis in all LCMT1 manipulation studies, as α4 appears to interact preferentially with unmethylated PP2Ac, potentially protecting it from degradation. Third, when interpreting phenotypes in LCMT1 knockout models, consider that effects may result from both decreased PP2A BAC holoenzymes and increased α4-PP2Ac complexes, necessitating additional controls that modulate α4 levels. Fourth, design rescue experiments that not only restore LCMT1 expression but also monitor redistribution of PP2Ac between various complexes. Fifth, implement proximity ligation assays to quantify PP2Ac interactions with different binding partners in situ, providing spatial context for complex redistribution. This approach addresses the observation that LCMT1 knockout generates a novel very high molecular weight PP2Ac-containing complex that appears to lack B subunit but contains α4 .

How should researchers design experiments to investigate potential therapeutic applications of modulating LCMT1 activity in hematopoietic disorders?

Designing experiments to investigate therapeutic applications of LCMT1 modulation requires a structured translational research approach. First, establish dose-dependent relationships between LCMT1 activity and hematopoietic parameters through genetic models with variable LCMT1 expression levels and small molecule modulators of methyltransferase activity . Second, develop high-throughput screening assays for compounds that enhance LCMT1 activity or inhibit the opposing methylesterase PME-1, focusing on molecules that increase PP2A methylation without cytotoxicity. Third, implement ex vivo expansion protocols for hematopoietic stem cells with LCMT1/PME-1 modulators, assessing both quantitative expansion and functional preservation through transplantation studies . Fourth, evaluate LCMT1 expression in patient samples across different hematopoietic disorders to identify conditions where LCMT1 modulation might be particularly beneficial. Fifth, conduct proof-of-concept studies in disease models using the most promising approaches, with careful assessment of off-target effects through phosphoproteomic analysis. This strategy builds on observations that enhancing LCMT1 function or inhibiting PME-1 may prove beneficial for expanding HSCs in vivo or ex vivo, with numerous potential clinical applications in hematopoietic disease .

What experimental approaches can distinguish between the effects of LCMT1 on PP2A, PP4, and PP6 methylation in cellular models?

Distinguishing between LCMT1 effects on different phosphatases requires sophisticated experimental approaches that isolate specific phosphatase complexes and their functions. First, develop and validate phosphatase-specific methylation-sensitive antibodies - existing antibodies can be pre-adsorbed with unmethylated PP2Ac C-terminal peptides to create antibodies specific for unmethylated PP4c, as demonstrated in previous research . Second, implement phosphatase-specific immunoprecipitation followed by methylation analysis using both antibody detection and mass spectrometry to quantify methylation levels of each phosphatase. Third, conduct comparative analyses of complex formation for each phosphatase using blue native PAGE, as research has shown LCMT1 loss differentially affects PP4 but not PP6 complex formation . Fourth, perform substrate-specific phosphatase activity assays using peptides or proteins preferentially dephosphorylated by individual phosphatases to determine functional consequences. Fifth, implement selective knockdown of each phosphatase catalytic subunit in LCMT1-manipulated cells to determine which phosphatase mediates specific phenotypes. These approaches address the complexity revealed by recent data showing LCMT1 loss dramatically reduces carboxyl methylation of PP4 and PP6 catalytic subunits and differentially affects formation of various phosphatase complexes .

What emerging technologies might enhance our understanding of LCMT1 function in cellular regulatory networks?

Emerging technologies offer unprecedented opportunities to elucidate LCMT1's role in cellular regulatory networks. CRISPR-based epigenetic modulation tools can enable precise temporal control of LCMT1 expression without permanent genetic modification, allowing researchers to distinguish between developmental and maintenance functions. Advanced proteomics approaches combining proximity labeling (BioID, TurboID) with LCMT1 fusion proteins will identify novel interaction partners and substrates beyond known phosphatases . Single-cell multi-omics technologies integrating transcriptomics, proteomics, and phosphoproteomics will reveal cell-type-specific LCMT1 functions and downstream effects, particularly relevant in heterogeneous systems like hematopoietic tissue. Cryo-electron microscopy of methylated versus unmethylated phosphatase complexes will provide structural insights into how methylation affects complex assembly and substrate recognition. Finally, phosphatase-specific biosensors based on fluorescence resonance energy transfer (FRET) will enable real-time visualization of phosphatase activity in living cells following LCMT1 modulation, bridging the gap between static biochemical analyses and dynamic cellular processes.

How might comparative studies across species inform our understanding of conserved LCMT1 functions?

Comparative studies across species can provide critical insights into evolutionarily conserved LCMT1 functions and species-specific adaptations. Begin by conducting phylogenetic analysis of LCMT1 sequence conservation across diverse organisms, from yeast (containing the homolog PPM1) to mammals, identifying conserved catalytic domains and regulatory regions . Implement cross-species complementation experiments to determine if LCMT1 from different species can rescue phenotypes in model organisms lacking endogenous LCMT1/PPM1. Compare phosphatase methylation patterns and the consequences of LCMT1/PPM1 disruption across species - research has shown disruption of PPM1 in yeast and LCMT1 in mammals both dramatically reduce PP2A BAC heterotrimers, demonstrating functional conservation . Examine species-specific differences in LCMT1 expression patterns, particularly across developmental stages and tissue types. Finally, conduct comparative structural biology studies of LCMT1-phosphatase interactions across species to identify conserved binding interfaces that may represent essential functional elements. This evolutionary perspective provides context for interpreting findings in model organisms and translating them to human biology.

What methodological approaches would best characterize the potential role of LCMT1 in non-canonical substrates beyond phosphatases?

Characterizing LCMT1's potential role with non-canonical substrates requires innovative methodological approaches that extend beyond traditional techniques. First, implement unbiased proteome-wide analyses of carboxyl methylation using antibodies against methylated C-terminal leucines combined with mass spectrometry to identify all cellular proteins bearing this modification . Second, develop LCMT1 substrate-trapping mutants that form stable complexes with potential substrates but cannot complete the methylation reaction, enabling identification of transient enzyme-substrate interactions. Third, conduct comparative methylome analysis between wild-type and LCMT1-deficient cells using methylation-specific enrichment strategies coupled with quantitative proteomics . Fourth, implement in vitro methyltransferase assays using purified LCMT1 and candidate substrate proteins identified through interactome studies. Fifth, validate identified substrates through site-directed mutagenesis of predicted methylation sites followed by functional studies to determine the physiological significance of these modifications. Finally, develop methylation-specific antibodies against newly identified substrates to enable direct visualization of methylation dynamics in cellular contexts.

How can conditional and tissue-specific LCMT1 knockout models advance our understanding beyond what global knockout studies have revealed?

Conditional and tissue-specific LCMT1 knockout models offer significant advantages over global knockout approaches for advancing our understanding of LCMT1 function. First, these models circumvent the embryonic lethality observed in global knockouts, enabling investigation of LCMT1 functions in adult tissues and specific developmental windows . Second, they allow discrimination between cell-autonomous effects and secondary consequences of LCMT1 loss by restricting deletion to specific cell types - particularly valuable for resolving whether LCMT1 has intrinsic or extrinsic roles in hematopoietic stem cell function . Third, tissue-specific models facilitate temporal studies by activating Cre-mediated recombination at defined timepoints, enabling dynamic assessment of immediate versus delayed effects of LCMT1 loss. Fourth, they permit detailed analysis of tissue-specific phosphatase complex distribution and function following LCMT1 deletion, addressing potential tissue-specific variations in the consequences of reduced methylation. Fifth, these models enable precise investigation of potential therapeutic applications by allowing targeted LCMT1 modulation in disease-relevant tissues without systemic effects. The gene trap knockout model generated in previous studies provides a foundation for developing these more sophisticated conditional systems .

Comparative Performance Metrics of Detection Methods Using Biotin-Conjugated LCMT1 Antibodies

Detection MethodSensitivity (Minimum Detectable LCMT1)Signal-to-Noise RatioAdvantagesLimitationsOptimal Application
Western Blot with Streptavidin-HRP10-20 ng8:1High specificity, quantifiableRequires cell lysisProtein expression quantification
Immunofluorescence with Streptavidin-Fluorophore50-100 ng/mL5:1Spatial information, multiplexing capabilityBackground in biotin-rich tissuesSubcellular localization studies
ELISA with Streptavidin-HRP5-10 ng/mL12:1High throughput, quantitativeNo spatial informationLarge-scale screening
Immunohistochemistry with Streptavidin-Peroxidase25-50 ng/mL7:1Tissue context, archival samplesReduced multiplexingClinical sample analysis
Flow Cytometry with Streptavidin-Fluorophore15-30 ng/mL6:1Single-cell resolution, multi-parameterLimited to suspendable cellsHematopoietic cell analysis

This comparative analysis is derived from experimental data using standardized samples and protocols . Sensitivity values represent approximate detection limits under optimal conditions. Signal-to-noise ratios were calculated based on specific signal intensity divided by background in negative control samples.

Effect of LCMT1 Knockout on PP2A Complex Formation and Related Phosphatases

Phosphatase ComplexReduction in LCMT1-/- vs WTMethylation DependenceAssociated PhenotypesDetection Method
PP2A BAC Heterotrimer92%HighEmbryonic lethality, hematopoietic defectsCo-IP, BN-PAGE
PP2A AC Core Dimer30%ModerateReduced steady-state levelsWestern blot
PP4 ComplexesVariable (complex-dependent)DifferentialNot fully characterizedCo-IP, BN-PAGE
PP6 ComplexesMinimalLowNot fully characterizedCo-IP, BN-PAGE
α4-PP2Ac ComplexIncrease (novel high MW complex)Inverse correlationUnknownBN-PAGE

Data compiled from experimental findings in LCMT1 knockout models demonstrates differential effects on various phosphatase complexes . LCMT1 loss causes a 92% reduction in PP2A BAC heterotrimers in whole embryos, with a 30% reduction in PP2A AC core dimers, indicating methylation is essential for complex formation and stability. PP4 complexes show variable methylation dependence, while PP6 complexes appear less affected. Notably, LCMT1 knockout increases formation of α4-PP2Ac complexes, suggesting a potential compensatory mechanism.

Optimization Parameters for Biotin-Conjugated LCMT1 Antibody Applications

ParameterWestern BlotImmunofluorescenceImmunohistochemistryELISAFlow Cytometry
Optimal Dilution1:500-1:20001:100-1:5001:200-1:10001:1000-1:50001:100-1:300
Incubation Time2h RT or overnight 4°COvernight 4°COvernight 4°C2h RT30-60 min on ice
Blocking Solution5% BSA in TBST10% normal serum + 1% BSA10% normal serum + avidin/biotin block1-3% BSA in PBS2% FBS in PBS
Antigen RetrievalN/ACitrate buffer pH 6.0, heatCitrate buffer pH 6.0, heatN/AN/A
Detection SystemStreptavidin-HRP (1:5000)Streptavidin-Alexa Fluor (1:1000)Streptavidin-HRP (1:1000)Streptavidin-HRP (1:10000)Streptavidin-PE (1:500)
Washing Steps3 × 5 min TBST3 × 10 min PBS-T3 × 5 min PBS-T4 × 3 min PBS-T2 × 3 min PBS

This optimization table provides starting parameters for different applications of biotin-conjugated LCMT1 antibodies based on experimental validation . Researchers should perform titration experiments to determine optimal conditions for their specific experimental system and sample type.

LCMT1 Expression and Function Across Developmental Stages in Hematopoietic Tissues

Developmental StageLCMT1 Expression LevelPP2A Methylation StatusHematopoietic Phenotype in LCMT1-/-Key Affected Cell Populations
E10.5-E12.5ModerateModerateMinor defects in early progenitorsEarly progenitors
E13.5-E14.5HighHighReduced HSC population, multilineage defectsKLS cells, erythroid progenitors
E15.5-E16.5Very HighVery HighSevere defects, typical lethalityAll lineages, KLS cells significantly reduced
E17.5+High (in surviving embryos)HighRare survivors show severe pancytopeniaLymphoid lineage most severely affected
Adult (conditional KO)Variable by tissueCorrelated with expressionPredicted HSC and progenitor exhaustionHSCs, multilineage effects

This developmental timeline is based on analysis of LCMT1 knockout studies in murine models . LCMT1 expression increases during critical periods of hematopoietic development, with peak expression corresponding to the timing of severe phenotypes in knockout models. Loss of LCMT1 causes progressive defects in hematopoiesis, affecting multiple lineages but with particular impact on KLS (c-Kit+, Lineage-, Sca-1+) cells, suggesting critical roles in HSC generation, self-renewal, and/or survival.

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.