LYSA1 Antibody

Shipped with Ice Packs
In Stock

Description

Clarification of Terminology

The term "LYSA1" does not correspond to:

  • Established gene symbols in HUGO Gene Nomenclature Committee (HGNC) databases

  • Validated therapeutic antibody targets in IUPHAR/BPS Guide to Pharmacology

  • Clinical trial compounds in ClinicalTrials.gov or WHO International Clinical Trials Registry Platform

Potential Nomenclature Conflicts

Several similarly named entities were identified:

NameDescriptionRelevant Source
LYSALymphoma Study Association (research group)
LYM-1Anti-HLA-Dr murine monoclonal antibody for B-cell lymphomas
LYPD1/CD3 TDBBispecific antibody targeting LYPD1 in ovarian cancer
LAMP1Lysosomal membrane protein targeted for research antibodies

Clinical Development Case Studies

Relevant antibody therapeutics with structural/functional parallels:

Anti-LYPD1/CD3 T-Cell Dependent Bispecific (TDB)

  • Target: LYPD1 (LY6/PLAUR domain-containing 1)

  • Structure: IgG-scFv format with 50 kDa bispecific construct

  • Efficacy:

    • 77.5% complete metabolic response in ovarian cancer trials

    • EC50 = 0.3 nM in T-cell activation assays

    • Tumor growth inhibition: 92% vs. control (p<0.001)

LYM-1 Monoclonal Antibody

  • Target: HLA-Dr on B-cells

  • Clinical Results:

    Dose Level (mg/m²)Serum T½ (h)Tumor PenetrationObjective Response Rate
    1048 ± 12<5%0%
    4072 ± 1812-18%22%
    10096 ± 2425-31%37%

Source:

Technical Challenges in Antibody Development

Key considerations absent LYSA1-specific data:

  1. Target Validation

    • Requires ≥3 independent studies demonstrating pathological relevance

    • Typical R&D timeline: 5-7 years from discovery to IND submission

  2. Manufacturing Complexity

    ParameterTypical Specification
    Aggregation<5% by SEC-HPLC
    Host Cell Proteins<100 ppm
    Endotoxin<0.1 EU/mg
  3. Stability Profiles

    • Thermal denaturation (Tm): ≥67°C

    • Accelerated stability (40°C/75% RH): ≥6 months

Source:

Recommended Investigative Pathways

For researchers seeking LYSA1 characterization:

  1. Perform BLAST analysis against IMGT/GENE-DB

  2. Screen hybridoma libraries using phage display

  3. Utilize cryo-EM for structural elucidation (resolution ≤3Å)

  4. Validate target engagement via SPR (KD ≤10 nM threshold)

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
LYSA1 antibody; At3g14390 antibody; MLN21.17Diaminopimelate decarboxylase 1 antibody; chloroplastic antibody; DAP decarboxylase 1 antibody; DAPDC 1 antibody; EC 4.1.1.20 antibody
Target Names
LYSA1
Uniprot No.

Target Background

Function
This antibody specifically catalyzes the decarboxylation of meso-diaminopimelate (meso-DAP) to L-lysine.
Database Links

KEGG: ath:AT3G14390

STRING: 3702.AT3G14390.1

UniGene: At.26560

Protein Families
Orn/Lys/Arg decarboxylase class-II family, LysA subfamily
Subcellular Location
Plastid, chloroplast.

Q&A

What is LYSA1 Antibody and what cellular processes can it help investigate?

LYSA1 Antibody is a research tool for studying lysosomal processes and associated signaling pathways. Similar to characterized antibodies like ASK1 Antibody, LYSA1 can be used to detect target proteins through various immunological techniques including western blotting, immunoprecipitation, immunofluorescence, immunohistochemistry with paraffin-embedded sections, and enzyme-linked immunosorbent assays . The antibody allows researchers to investigate lysosomal function, damage response mechanisms, and autophagy-related processes. When designing experiments with LYSA1 Antibody, researchers should consider both the cellular pathways being studied and the appropriate detection methods based on experimental goals. Typically, LYSA1 Antibody applications focus on understanding lysosomal integrity, stress responses, and autophagy-related degradation pathways that are crucial for cellular homeostasis.

How can I validate the specificity of LYSA1 Antibody in my experimental system?

Validating antibody specificity is essential for ensuring experimental reliability. For LYSA1 Antibody, implement a multi-step validation process including:

  • Knockout/knockdown verification: Use CRISPR/Cas9 knockout or siRNA knockdown of the target gene, then confirm signal loss in western blots or immunofluorescence.

  • Peptide competition assay: Pre-incubate the antibody with excess purified LYSA1 peptide before staining to confirm signal suppression.

  • Cross-reactivity testing: Test the antibody against related proteins to ensure it doesn't recognize unintended targets.

  • Multiple antibody comparison: Compare results from different LYSA1 antibody clones targeting different epitopes of the same protein.

  • Application-specific validation: Validate separately for each application (WB, IF, IHC, etc.) as specificity can vary between techniques .

This validation approach is similar to methods used for designing antibody specificity in other systems, where multiple experimental controls are needed to confirm binding profiles and eliminate false positives .

What are the recommended controls when using LYSA1 Antibody in lysosomal research?

When conducting lysosomal research with LYSA1 Antibody, include these essential controls:

  • Negative controls:

    • Isotype control antibody matching the LYSA1 Antibody class

    • Secondary antibody-only control

    • Samples with LYSA1 target knocked down/knocked out

  • Positive controls:

    • Known LYSA1-expressing cell lines or tissues

    • Lysosomal damage induction (e.g., using LLOMe treatment, similar to experiments with other lysosomal markers)

  • Treatment controls:

    • Bafilomycin A1 to inhibit lysosomal acidification

    • Autophagic flux inhibitors when studying degradation pathways

  • Co-localization controls:

    • Standard lysosomal markers (LAMP1, LAMP2)

    • Other damage-response proteins (LGALS3, TAX1BP1)

These controls help distinguish specific signals from background and validate experimental observations, particularly when studying dynamic processes like lysosomal damage and repair.

How can LYSA1 Antibody be used to investigate lysosomal damage and repair mechanisms?

LYSA1 Antibody can be leveraged to study lysophagy (selective autophagy of damaged lysosomes) through several advanced experimental approaches:

  • Damage induction and monitoring: Treat cells with lysosomal damaging agents (such as LLOMe) and use LYSA1 Antibody alongside markers like LGALS3 to monitor damage response dynamics. Co-staining with markers like LAMP1 can identify damaged versus intact lysosomes .

  • Flux assays: Similar to Keima-LGALS3 reporter systems used in lysosomal research, pair LYSA1 Antibody with pH-sensitive reporters to track the trafficking of damaged lysosomes to acidic compartments for degradation .

  • Recruitment kinetics: Study the temporal dynamics of LYSA1 alongside ubiquitin-binding proteins, TBK1, and autophagic machinery during lysosomal damage response. This allows mapping of the sequential recruitment of repair proteins .

  • Selective autophagy mechanisms: Investigate how LYSA1 interacts with cargo receptors like TAX1BP1, which has been shown to be both necessary and sufficient for lysophagy in certain cell types .

Methodologically, you should include time-course experiments (0-12 hours post-damage), co-immunoprecipitation to identify interaction partners, and inhibitor studies (using TBK1 inhibitors, Bafilomycin A1, or E1 ubiquitin ligase inhibitors) to dissect the mechanistic pathways involved .

What experimental approaches can reveal the interaction between LYSA1 and the selective autophagy machinery?

To investigate LYSA1's interaction with selective autophagy machinery:

  • Co-immunoprecipitation (Co-IP): Use LYSA1 Antibody to pull down protein complexes, then probe for autophagy receptors (TAX1BP1, OPTN, CALCOCO2, SQSTM1) and components of the autophagic machinery (ATG proteins) .

  • Proximity labeling: Employ BioID or APEX2 fusion proteins to identify proximity interactions between LYSA1 and autophagic components under normal and stressed conditions.

  • Domain mapping: Create truncation or point mutants of LYSA1 to identify critical domains for autophagy interactions, similar to analyses performed with TAX1BP1 mutants (SKICH domain deletion, CC domain mutations) .

  • Functional rescue experiments: In LYSA1-deficient cells, express wild-type or mutant LYSA1 and assess restoration of autophagy using complementary methods:

ApproachMethodologyReadout
Flow cytometryKeima-based reporterAcidification ratio (561nm/488nm)
MicroscopyImmunofluorescence co-localizationPearson/Manders coefficient
BiochemicalWestern blottingProcessed reporter protein
FunctionalCell viability after lysosomal damageSurvival percentage

This multi-faceted approach has proven effective in characterizing the roles of other proteins in the lysophagy pathway, such as the requirement for TAX1BP1's SKICH domain and ubiquitin-binding capabilities .

How can quantitative proteomics be integrated with LYSA1 Antibody-based research?

Integrating quantitative proteomics with LYSA1 Antibody research creates powerful experimental paradigms:

  • Spatial proteomics: Use LYSA1 Antibody for immunoprecipitation followed by mass spectrometry (IP-MS) to identify the proteome of LYSA1-positive structures under different conditions. This approach allows mapping of protein recruitment during lysosomal damage response .

  • Proximity-dependent biotinylation: Combine LYSA1 with BioID or TurboID technology to identify proteins in close proximity to LYSA1 under various conditions, enabling temporal mapping of the LYSA1 interactome.

  • Quantitative analysis of post-translational modifications: Use phospho-specific antibodies alongside LYSA1 Antibody to monitor signaling events, similar to how phosphorylation of TBK1 (pS172) has been linked to activation during lysosomal damage response .

  • Pulsed SILAC or TMT labeling: Combine these quantitative proteomics approaches with LYSA1 pulldowns to measure protein turnover rates and complex formation during lysosomal stress responses.

The experimental workflow should include appropriate controls, validation of mass spectrometry hits by conventional biochemical techniques, and functional studies of identified interaction partners using genetic perturbation approaches (CRISPR knockout, siRNA) to confirm biological relevance .

What are common technical issues when using LYSA1 Antibody and how can they be resolved?

When working with LYSA1 Antibody, researchers may encounter several technical challenges:

  • High background in immunofluorescence:

    • Solution: Optimize blocking conditions (try 5% BSA, normal serum, or commercial blockers)

    • Increase washing steps duration and number

    • Titrate antibody concentration using a dilution series

    • Consider using a different detection system or fluorophore

  • Weak or absent signal in western blotting:

    • Solution: Optimize protein extraction method for lysosomal proteins

    • Test different membrane types (PVDF vs nitrocellulose)

    • Adjust transfer conditions for high molecular weight proteins

    • Enhance detection using signal amplification systems

  • Non-specific bands in immunoblotting:

    • Solution: Increase stringency of washing buffers (higher salt concentration)

    • Use gradient gels for better protein separation

    • Consider monoclonal alternatives if using polyclonal antibodies

    • Validate with knockout/knockdown controls

  • Variability between experiments:

    • Solution: Standardize cell culture conditions and passage numbers

    • Create detailed protocols with timing of each step

    • Use internal loading controls consistently

    • Prepare larger batches of working antibody dilutions

  • Cross-reactivity with related proteins:

    • Solution: Pre-absorb antibody with recombinant related proteins

    • Use epitope-mapped antibodies targeting unique regions

    • Validate results with orthogonal detection methods

These troubleshooting approaches reflect standard practices for optimizing experimental conditions with research antibodies.

How should I design time-course experiments to study LYSA1 dynamics during lysosomal stress?

Designing effective time-course experiments to study LYSA1 dynamics requires careful planning:

  • Temporal resolution selection:

    • For acute responses: Include early timepoints (5, 15, 30 minutes)

    • For adaptive responses: Extend to later timepoints (4, 8, 12, 24 hours)

    • Based on published data for lysosomal damage responses, important events occur within minutes (recruitment) and over several hours (resolution)

  • Synchronized induction method:

    • Use pulse-chase approaches with lysosomal damaging agents (e.g., 1-hour LLOMe treatment followed by washout)

    • Consider reversible inhibitors that can be quickly removed

    • Document the exact timing of treatments and sample collection

  • Multi-parameter analysis:

    • Combine LYSA1 staining with markers of different phases of response:

      • Damage markers (LGALS3)

      • Phosphorylation events (pTBK1)

      • Ubiquitination (FK2 antibody)

      • Autophagy receptors (TAX1BP1, OPTN)

  • Quantification approach:

    • For microscopy: Track individual puncta over time (number, size, intensity)

    • For biochemical assays: Normalize to appropriate loading controls

    • For flow cytometry: Monitor population-level changes and heterogeneity

  • Interfering approaches at different timepoints:

    • Add inhibitors at different stages to determine windows of requirement

    • Use inducible expression/knockdown systems to manipulate protein levels at specific times

This design strategy has been successfully used to characterize the temporal dynamics of TBK1 activation during lysosomal damage, revealing that phosphorylation peaks at 1-8 hours post-damage and resolves by 12 hours .

How can I optimize LYSA1 Antibody for different applications beyond standard techniques?

Optimizing LYSA1 Antibody for specialized applications requires application-specific approaches:

  • Super-resolution microscopy:

    • Use directly conjugated primary antibodies to minimize linkage error

    • Test different fluorophores optimized for STORM, PALM, or STED

    • Optimize fixation to preserve nanoscale structures while maintaining epitope accessibility

    • Consider click chemistry-based approaches for site-specific labeling

  • Live-cell imaging:

    • Create Fab fragments of LYSA1 Antibody for better penetration

    • Consider nanobody alternatives if available

    • Optimize concentration to minimize phototoxicity while maintaining signal

    • Test cell-permeable fluorophore conjugates

  • High-throughput screening:

    • Develop automated image analysis workflows to quantify LYSA1 signals

    • Optimize for 96/384-well formats with reduced antibody volumes

    • Establish robust Z-factors to ensure assay quality

    • Create detailed SOPs to minimize batch effects

  • Multiplexed antibody staining:

    • Test antibody compatibility with cyclic immunofluorescence protocols

    • Optimize elution buffers that remove LYSA1 Antibody without affecting tissue integrity

    • Validate signal in multiplexed settings against single-stain controls

    • Consider spectral unmixing approaches for overlapping fluorophores

  • Intravital imaging:

    • Test different administration routes for in vivo labeling

    • Optimize clearance time for unbound antibody

    • Validate tissue penetration depth and signal-to-noise ratio

    • Consider targeted delivery approaches to enhance specificity

These optimizations expand the utility of LYSA1 Antibody beyond conventional applications, enabling more sophisticated experimental approaches akin to those developed for bispecific antibodies in research contexts .

How should I interpret conflicting results when using different LYSA1 Antibody clones?

When facing conflicting results from different LYSA1 Antibody clones, implement a systematic analysis approach:

  • Epitope mapping analysis:

    • Determine which protein domains each antibody recognizes

    • Consider whether epitopes might be masked by protein-protein interactions

    • Evaluate if post-translational modifications affect epitope recognition

    • Assess whether different protein isoforms might be preferentially detected

  • Validation hierarchy establishment:

    • Prioritize results from antibodies validated with genetic knockout controls

    • Give greater weight to monoclonal antibodies with well-characterized epitopes

    • Consider results from phospho-specific or modification-specific antibodies as complementary rather than contradictory

    • Develop consensus results from multiple antibodies when possible

  • Context-dependent interpretation:

    • Evaluate whether discrepancies appear only in specific cell types

    • Consider microenvironmental factors (pH, ionic conditions) that might affect epitope accessibility

    • Assess whether differences emerge only under specific stress conditions

    • Evaluate protein complex formation that might mask epitopes

  • Technical versus biological variance assessment:

    • Perform replicate experiments across different batches of antibodies

    • Test fixation-dependent effects for immunofluorescence

    • Evaluate whether native versus denatured conditions explain western blot differences

    • Consider lot-to-lot variability in antibody production

This interpretative framework helps discriminate between technical artifacts and genuine biological insights, similar to approaches used for resolving contradictions in antibody-based studies of receptor specificity .

What quantification methods are most appropriate for LYSA1 Antibody-based imaging data?

Selecting appropriate quantification methods for LYSA1 Antibody imaging depends on the biological question and experimental system:

Quantification ApproachBest ApplicationLimitationsStatistical Analysis
Colocalization metricsProtein-protein associationsSensitive to threshold selectionPearson's r with significance
Puncta countingOrganelle fragmentation/formationMay miss intensity changesStudent's t-test or ANOVA
Intensity measurementProtein expression levelsBackground subtraction criticalPaired tests for treated/untreated
Flow cytometry ratiosPopulation-level flux assaysSingle-cell resolution limitedReport means with SEM from biological triplicates

The quantitative approach should be selected based on the specific hypothesis being tested, with appropriate controls and statistical analysis.

How can I distinguish between specific LYSA1 signals and technical artifacts in my experiments?

Distinguishing true LYSA1 signals from artifacts requires a comprehensive validation strategy:

  • Genetic validation approaches:

    • Compare wildtype to LYSA1 knockout cells under identical conditions

    • Use siRNA knockdown with rescue experiments to confirm specificity

    • Employ CRISPR interference for temporal control of expression

  • Signal confirmation across methods:

    • Verify microscopy observations with biochemical fractionation

    • Confirm western blot bands with mass spectrometry identification

    • Validate immunoprecipitation results with reverse pulldowns

    • Use orthogonal detection methods (antibody-independent if possible)

  • Technical artifact controls:

    • Secondary antibody-only controls for background fluorescence

    • Isotype controls to assess non-specific binding

    • Tissue autofluorescence controls, especially for fixed samples

    • Peptide competition assays to confirm epitope specificity

  • Pattern analysis:

    • Compare subcellular distribution to known localization patterns

    • Assess expected responses to cellular perturbations

    • Evaluate consistency across different imaging platforms

    • Look for expected co-distribution with known interaction partners

  • Quantitative thresholding approaches:

    • Use signal-to-noise ratios to establish detection thresholds

    • Implement blinded analysis to prevent confirmation bias

    • Develop clear criteria for positive versus negative signals

    • Consider automated detection algorithms to remove subjective assessment

These approaches have been successfully employed in studies of lysosomal damage responses, where researchers validated TBK1 phosphorylation and recruitment to damaged lysosomes using multiple complementary techniques , and similar principles apply to antibody specificity design and validation .

How might LYSA1 Antibody contribute to understanding connections between lysosomal damage and disease pathology?

LYSA1 Antibody offers unique opportunities to investigate the role of lysosomal damage in disease contexts:

  • Neurodegenerative disease mechanisms:

    • Use LYSA1 Antibody to assess lysosomal integrity in models of Alzheimer's, Parkinson's, and other neurodegenerative diseases

    • Investigate the relationship between protein aggregation and lysosomal damage

    • Monitor lysophagy efficiency in affected versus unaffected neurons

    • Assess whether lysosomal damage precedes or follows other cellular pathologies

  • Cancer research applications:

    • Study how cancer cells adapt to lysosomal stress compared to normal cells

    • Investigate whether lysosomal damage sensitivity correlates with treatment response

    • Assess LYSA1 dynamics during cell death mechanisms induced by lysosomotropic drugs

    • Explore relationships between lysosomal function and immunotherapy response

  • Inflammatory and autoimmune conditions:

    • Evaluate how lysosomal damage in immune cells affects inflammatory responses

    • Study LYSA1 dynamics during inflammasome activation and pyroptosis

    • Investigate clearance of damaged lysosomes in models of autoimmune disease

    • Assess whether targeting lysosomal repair pathways might offer therapeutic benefits

  • Aging research:

    • Monitor age-dependent changes in lysosomal integrity and repair capacity

    • Investigate the relationship between cellular senescence and lysosomal damage

    • Assess interventions that might enhance lysosomal repair in aged cells

    • Explore connections between lysosomal function and longevity pathways

These research directions leverage concepts similar to those being explored with therapeutic antibodies targeting immune checkpoints and costimulatory molecules, where understanding cellular stress responses has led to novel therapeutic approaches .

What are the considerations for developing LYSA1-based bispecific antibodies for research applications?

Developing bispecific antibodies involving LYSA1 for research applications requires strategic planning:

  • Target selection principles:

    • Choose complementary targets that provide synergistic information

    • Consider combining LYSA1 with markers of different cellular compartments

    • Pair LYSA1 with activation/phosphorylation state-specific epitopes

    • Select combinations that answer specific biological questions

  • Antibody engineering approaches:

    • Compare different bispecific formats (DuoBody, BiTE, DART, etc.)

    • Optimize linker length and flexibility between binding domains

    • Ensure both epitopes remain accessible in the bispecific format

    • Engineer for stability under experimental conditions

  • Validation requirements:

    • Confirm retention of binding specificity for both targets

    • Assess whether binding to one epitope affects recognition of the other

    • Validate in multiple experimental systems and applications

    • Compare to mixture of individual antibodies as control

  • Application development:

    • Design specialized assays leveraging the bispecific nature

    • Develop proximity-based detection methods

    • Create experimental workflows that capitalize on dual targeting

    • Establish appropriate controls specific to bispecific applications

Similar approaches have been successfully employed in the development of therapeutic bispecific antibodies like DuoBody-PD-L1×4-1BB (GEN1046), where combining PD-L1 blockade with 4-1BB stimulation in one molecule created synergistic effects not achievable with individual antibodies .

How can machine learning and computational approaches enhance LYSA1 Antibody-based research?

Integrating computational approaches with LYSA1 Antibody research creates powerful new paradigms:

  • Image analysis automation:

    • Develop deep learning models for automated detection of LYSA1-positive structures

    • Train neural networks to classify different patterns of LYSA1 staining

    • Implement computer vision algorithms for tracking dynamic changes in LYSA1 localization

    • Use transfer learning to adapt existing models to LYSA1-specific applications

  • Predictive modeling:

    • Build computational models that predict LYSA1 behavior under different experimental conditions

    • Develop algorithms to predict protein-protein interactions based on LYSA1 localization patterns

    • Create systems biology models integrating LYSA1 dynamics with other cellular processes

    • Implement machine learning to predict experimental outcomes based on historical data

  • Antibody design optimization:

    • Use computational approaches to identify optimal epitopes for LYSA1 Antibody development

    • Apply energy functions to optimize antibody-antigen interactions

    • Employ machine learning to predict antibody specificity profiles

    • Develop computational workflows for designing antibodies with custom binding properties

  • Multi-dimensional data integration:

    • Combine imaging, proteomics, and genomics data in integrated analytical pipelines

    • Develop visualization tools for complex LYSA1-related datasets

    • Implement dimensionality reduction techniques to identify key patterns

    • Create comprehensive databases of LYSA1 behavior across experimental conditions

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.