GARS Antibody

Glycyl-TRNA Synthetase, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Function of GARS Antibody

GARS (glycyl-tRNA synthetase) antibody is a research tool designed to detect and quantify the GARS protein, a class II aminoacyl-tRNA synthetase responsible for attaching glycine to its cognate tRNA. This enzyme also synthesizes diadenosine tetraphosphate (Ap4A), a signaling molecule involved in cellular regulation . The antibody facilitates studies on GARS’s role in protein synthesis, cellular stress responses, and its implications in diseases like cancer and neurodegenerative disorders .

Types and Applications of GARS Antibodies

GARS antibodies are categorized by host species, clonality, and application suitability. Below is a comparative analysis of commercially available antibodies:

AntibodyHostClonalityApplicationsReactivityDilution (WB)Source
ab125008 (Abcam)RabbitMonoclonalIHC-P, WBHuman, Mouse, RatN/A
CAB13305 (Assay Genie)RabbitPolyclonalWBHuman1:2000
ab282154 (Abcam)MouseMonoclonalICC, WBHumanN/A
67893-1-Ig (Proteintech)MouseMonoclonalWB, IHC, ICC, IFHuman, Mouse, Rat1:5000–1:50000
ab196496 (Abcam)RabbitPolyclonalWB, IHC-P, ICC/IFHuman, Mouse, RatN/A
15831-1-AP (Proteintech)RabbitPolyclonalWB, IHC, IF, IP, RIPHuman, Mouse, Rat1:5000–1:50000

Key Applications:

  • Western Blot (WB): Quantitative analysis of GARS expression in cell lysates .

  • Immunohistochemistry (IHC): Localization of GARS in tissue sections, particularly in cancer studies .

  • Immunofluorescence (ICC/IF): Subcellular localization in cultured cells .

  • Immune Precipitation (IP): Protein interaction studies .

Role in Cancer Prognosis

GARS overexpression correlates with poor prognosis in multiple cancers:

  • Bladder Cancer (BC): High GARS levels in tumor tissues and urinary extracellular vesicles (uEVs) predict aggressive disease and metastasis . Knockdown of GARS inhibits cell proliferation and migration .

  • Prostate Cancer (PCa): GARS expression increases with Gleason score progression, associating with nodal metastasis and residual tumor presence .

  • Pan-Cancer Immunotherapy: GARS1 expression predicts response to anti-PD-L1 therapy and correlates with tumor mutational burden (TMB) and neoantigen load .

Mechanistic Insights

  • Immune Microenvironment: GARS upregulation correlates with infiltration of neutrophils, macrophages, and CD8+ T cells in tumors, suggesting dual roles in immune suppression and activation .

  • Drug Sensitivity: GARS-high tumors show sensitivity to ifosfamide, auranofin, DMAPT, and A-1331852, highlighting therapeutic potential .

  • Epithelial-Mesenchymal Transition (EMT): GARS promotes cell migration and invasion via pathways linked to MYC/E2F targets .

Diagnostic and Therapeutic Potential

AspectDetailsReferences
BiomarkerGARS detection in uEVs offers a non-invasive method for BC prognosis .
Therapeutic TargetInhibiting GARS may reduce cancer cell proliferation and metastasis .
ImmunotherapyGARS expression predicts anti-PD-L1 therapy efficacy .

Product Specs

Introduction
Glycyl-tRNA synthetase (GARS) is an enzyme that belongs to the class II family of tRNA synthetases. As a member of the aminoacyl-tRNA synthetase family, GARS plays a crucial role in protein synthesis by catalyzing the attachment of glycine to its corresponding tRNA molecule, tRNA(Gly). Beyond its role in protein synthesis, GARS also exhibits an intriguing ability to produce diadenosine tetraphosphate (Ap4A). Ap4A is a molecule with diverse regulatory functions, acting as a universal signaling molecule in various cellular processes. The production of Ap4A by GARS occurs through the direct condensation of two ATP molecules. Interestingly, GARS has been identified as a target of autoantibodies in certain autoimmune diseases, specifically polymyositis and dermatomyositis. The presence of these autoantibodies highlights the potential involvement of GARS dysregulation in the pathogenesis of these conditions.
Physical Appearance
A clear and colorless solution that has been sterilized through filtration.
Formulation
The antibody is supplied at a concentration of 1 milligram per milliliter (mg/ml) in a buffer solution containing phosphate-buffered saline (PBS) at a pH of 7.4, 10% glycerol, and 0.02% sodium azide.
Storage Procedures
For short-term storage of up to one month, the antibody should be kept at a refrigerated temperature of 4 degrees Celsius. To ensure optimal long-term storage, it is recommended to store the antibody at a temperature of -20 degrees Celsius. Repeated cycles of freezing and thawing should be avoided.
Stability / Shelf Life
When stored properly at -20°C, the antibody remains stable for a period of 12 months. At a refrigerated temperature of 4°C, the antibody can be stored for up to 1 month.
Applications
The GARS antibody has undergone rigorous testing using ELISA (Enzyme-Linked Immunosorbent Assay) and Western blot analysis. These tests confirm the antibody's specificity for its target protein and its ability to bind with high affinity. While the antibody has been validated for these applications, it is important to note that optimal working dilutions may vary depending on the specific experimental conditions. Therefore, it is recommended to perform a titration experiment to determine the most suitable dilution for each individual assay. A starting dilution of 1:500 is a good initial point for optimization.
Synonyms
Glycine--tRNA ligase, Diadenosine tetraphosphate synthetase, AP-4-A synthetase, Glycyl-tRNA synthetase, GlyRS, GARS, CMT2D, DSMAV, HMN5, SMAD1.
Purification Method
GARS antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT4E10AT.
Immunogen
Anti-human GARS mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human GARS amino acids 43-289 purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is GARS and why are GARS antibodies significant in research?

GARS (glycyl-tRNA synthetase) is an aminoacyl-tRNA synthetase that catalyzes the ATP-dependent ligation of glycine to the 3'-end of its cognate tRNA. It functions as an (alpha)2 dimer belonging to the class II family of tRNA synthetases . Beyond its canonical role in protein synthesis, GARS produces diadenosine tetraphosphate (Ap4A), a universal pleiotropic signaling molecule needed for cell regulation pathways .

GARS antibodies have become crucial research tools for several reasons:

  • They enable investigation of both canonical aminoacylation functions and non-canonical signaling roles

  • They facilitate research on GARS-associated diseases, including neurodegenerative disorders such as Distal Hereditary Motor Neuronopathy and Spinal Muscular Atrophy

  • They allow exploration of protein-protein interactions within the multi-tRNA synthetase complex

  • They help examine subcellular localization patterns in different tissues and cell types

  • They provide validation for gene manipulation experiments including knockdown or knockout studies

With increasing interest in aminoacyl-tRNA synthetases as potential therapeutic targets, GARS antibodies have become particularly valuable for drug discovery research and understanding fundamental biological processes .

What are the validated applications for GARS antibodies?

GARS antibodies serve as versatile research tools utilized across multiple molecular and cellular biology techniques. Based on extensive validation data, GARS antibodies can be effectively employed in:

ApplicationDescriptionValidated SamplesReference
Western Blot (WB)Detection of GARS protein (75-80 kDa)Human brain tissue, HeLa cells, HepG2 cells, HT-1080 cells
Immunohistochemistry (IHC)Tissue distribution analysisHuman skin cancer tissue, mouse brain tissue
Immunofluorescence (IF)/ICCSubcellular localization studiesHepG2 cells
Immunoprecipitation (IP)Isolation of GARS complexesMouse liver tissue
Co-immunoprecipitation (CoIP)Protein-protein interaction studiesMultiple cell types
RNA immunoprecipitation (RIP)RNA-protein interaction analysisVarious experimental systems
ELISAQuantitative detection in solutionMultiple experimental systems

The selection of appropriate application depends on your specific research question, available samples, and experimental design requirements. Each application requires specific optimization for maximum sensitivity and specificity.

How do I select between polyclonal and monoclonal GARS antibodies?

The choice between polyclonal and monoclonal GARS antibodies significantly impacts experimental outcomes and should be based on specific research requirements:

Polyclonal GARS Antibodies:

  • Recognize multiple epitopes on the GARS protein, increasing detection sensitivity

  • Provide robust signal in applications like WB and IHC

  • Examples include rabbit polyclonal antibodies such as TA363030 (OriGene) and 15831-1-AP (Proteintech)

  • Best suited for: initial characterization studies, detection of denatured proteins, and applications requiring high sensitivity

Monoclonal GARS Antibodies:

  • Recognize a single epitope on the GARS protein

  • Offer superior specificity and batch-to-batch consistency

  • Produce lower background and reduced cross-reactivity

  • Best suited for: reproducible experiments, specific epitope targeting, and quantitative analyses

Selection criteria should include:

  • Application requirements (polyclonals often perform better in IHC, while monoclonals may be preferred for therapeutic or diagnostic applications)

  • Required species reactivity (verify if the antibody recognizes GARS from your species of interest)

  • Epitope location importance (N-terminal, C-terminal, or internal regions may be differentially accessible in various applications)

  • Available validation data for your specific application and model system

For critical research applications, validating antibody performance in your specific experimental system is recommended regardless of the antibody type selected.

What dilution factors are optimal for different GARS antibody applications?

Optimal dilution factors for GARS antibodies vary by application, antibody affinity, and sample type. Based on validated protocols, the following dilution ranges are recommended:

ApplicationRecommended Dilution RangeOptimization ConsiderationsReference
Western Blot (WB)1:500-1:2000Start with 1:1000 and adjust based on signal-to-noise ratio
Immunohistochemistry (IHC)1:50-1:500Lower dilutions for FFPE tissues, higher for frozen sections
Immunofluorescence (IF)/ICC1:10-1:100Begin with 1:50 for cultured cells
Immunoprecipitation (IP)0.5-4.0 μg per 1.0-3.0 mg total proteinAntibody:protein ratio is more critical than dilution factor
ELISAVariableRequires systematic optimization for each assay format

Important considerations for dilution optimization include:

  • Always perform titration experiments with serial dilutions to determine optimal concentration for your specific system

  • Evaluate signal-to-noise ratio rather than absolute signal strength

  • Different tissues and cell types may require adjusted antibody concentrations based on GARS expression levels

  • More sensitive detection systems (chemiluminescence, fluorescence) allow higher dilutions than colorimetric methods

  • Documentation of optimization conditions is essential for experimental reproducibility

As noted in product literature, "It is recommended that this reagent should be titrated in each testing system to obtain optimal results" as optimal dilution can be "Sample-dependent" .

How can GARS antibodies be optimized for co-immunoprecipitation studies?

Optimizing GARS antibodies for co-immunoprecipitation (CoIP) studies requires careful consideration of multiple experimental parameters:

  • Antibody Selection:

    • Choose antibodies specifically validated for CoIP applications

    • Consider antibodies targeting different GARS epitopes to avoid interfering with protein-protein interaction sites

    • Polyclonal antibodies often perform well in CoIP due to their recognition of multiple epitopes

  • Lysate Preparation:

    • Use mild lysis buffers (e.g., NP-40 or Triton X-100 based) to preserve protein-protein interactions

    • Include protease and phosphatase inhibitor cocktails to prevent degradation

    • Optimize protein concentration (typically 1.0-3.0 mg of total protein lysate recommended)

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

  • Antibody Concentration and Incubation:

    • Use optimal antibody amounts (typically 0.5-4.0 μg for GARS antibodies)

    • Allow sufficient incubation time (overnight at 4°C is generally effective)

    • Use gentle rotation to maintain antibody-antigen contact while minimizing disruption of complexes

  • Essential Controls:

    • Include IgG isotype control to assess non-specific binding

    • If available, include GARS knockout/knockdown samples as negative controls

    • Consider reverse CoIP (immunoprecipitating with antibodies against suspected interacting partners)

    • Include input samples (pre-immunoprecipitation lysate) for comparison

  • Detection Methods:

    • For unbiased identification of interacting partners, use mass spectrometry

    • For known interactions, western blotting with specific antibodies against suspected partners

    • Consider protein crosslinking approaches for capturing transient interactions

Research has demonstrated that antibodies targeting individual members of the multi-tRNA synthetase complex can successfully detect all members of the complex co-immunoprecipitating with the target across several cell types . This approach has effectively validated recombinant antibodies against aminoacyl-tRNA synthetases, including GARS.

How can GARS antibodies help investigate non-canonical functions of glycyl-tRNA synthetase?

GARS antibodies provide valuable tools for investigating emerging non-canonical functions beyond the enzyme's primary role in protein synthesis:

  • Subcellular Localization Studies:

    • Immunofluorescence with GARS antibodies can track non-canonical localization patterns

    • Combination with organelle markers identifies unexpected cellular compartments where GARS may function

    • Comparison of localization under different cellular conditions (stress, disease states) reveals context-dependent functions

  • Protein-Protein Interaction Analysis:

    • Co-immunoprecipitation with GARS antibodies followed by mass spectrometry identifies novel interaction partners outside the tRNA synthetase complex

    • Validation using reverse co-immunoprecipitation and proximity ligation assays confirms specific interactions

    • Investigation of condition-specific interactions reveals regulatory mechanisms

  • Ap4A Signaling Pathway Investigation:

    • GARS produces diadenosine tetraphosphate (Ap4A), a universal pleiotropic signaling molecule

    • Immunoprecipitation of GARS followed by measurement of associated Ap4A production quantifies this non-canonical activity

    • Analysis of correlation between GARS localization, interaction partners, and Ap4A production reveals regulatory mechanisms

  • Disease-Associated Variant Analysis:

    • Comparison of wild-type and mutant GARS using antibodies that recognize both forms

    • Investigation of how disease mutations specifically affect non-canonical functions, including altered interactions or localization

    • Correlation of biochemical findings with clinical phenotypes

  • Post-translational Modification Mapping:

    • Immunoprecipitation with GARS antibodies followed by mass spectrometry identifies post-translational modifications

    • Correlation of modifications with non-canonical functions or altered localization patterns

    • Development of modification-specific antibodies to track regulated forms of GARS

By combining these approaches with genetic manipulation techniques and appropriate controls, researchers can effectively distinguish canonical aminoacylation functions from emerging non-canonical roles in signaling, regulation, and disease pathogenesis.

What validation methods are essential for confirming GARS antibody specificity?

Thorough validation of GARS antibody specificity is crucial for obtaining reliable research results. The following comprehensive validation methods are recommended:

  • Genetic Validation Approaches:

    • GARS knockdown/knockout testing: Evaluate antibody on samples with reduced or eliminated GARS expression

    • Overexpression validation: Test on samples with overexpressed GARS (wild-type or tagged variants)

    • Correlation of signal intensity with known expression levels across sample types

  • Multiple Antibody Strategy:

    • Employ multiple antibodies targeting different GARS epitopes

    • Consistent results across different antibodies significantly increase confidence in specificity

    • Compare commercially available antibodies from different sources (e.g., 15831-1-AP from Proteintech and TA363030 from OriGene )

  • Immunoblot Analytical Parameters:

    • Verify single band at expected molecular weight (75-80 kDa for GARS)

    • Confirm absence of non-specific bands across multiple sample types

    • Include appropriate positive controls (tissues/cells known to express GARS, such as human brain tissue, HeLa cells)

    • Include negative controls (tissues/cells with minimal GARS expression)

  • Immunoprecipitation-Mass Spectrometry:

    • Perform IP with the GARS antibody followed by mass spectrometry analysis

    • Confirm GARS as the predominant protein identified in the immunoprecipitate

    • This approach has been successfully employed to validate recombinant antibodies against aminoacyl-tRNA synthetases

  • Peptide Competition Assay:

    • Pre-incubate antibody with the immunizing peptide (if available)

    • Verify signal reduction or elimination in applications like WB or IHC

    • This confirms epitope-specific binding rather than non-specific interactions

  • Cross-Application Validation:

    • Test antibody performance across multiple applications (WB, IHC, IF, IP)

    • Consistent results across different methodologies substantially increase confidence in specificity

    • Document optimization conditions for each application systematically

Implementing multiple validation approaches provides the strongest evidence for antibody specificity. Thorough documentation of these validation steps is increasingly required by leading journals and funding agencies for research publication and reproducibility.

How are GARS antibodies used in studying neurodegenerative disorders?

GARS antibodies serve as essential tools in studying neurodegenerative disorders, particularly those linked to mutations in the GARS1 gene, including Distal Hereditary Motor Neuronopathy and Spinal Muscular Atrophy . These antibodies enable several critical research approaches:

  • Mutation-Specific Analyses:

    • Detection of wild-type versus mutant GARS protein expression patterns

    • Comparative analysis of protein stability and half-life between normal and disease-associated variants

    • Examination of potential conformational changes in mutant proteins using epitope accessibility studies

  • Subcellular Localization Studies:

    • Immunofluorescence with GARS antibodies to track mislocalization of mutant GARS in neuronal cells

    • Co-localization analysis with neuronal markers in neurodegeneration models

    • Time-course studies monitoring GARS distribution during disease progression

  • Protein Interaction Network Analysis:

    • Immunoprecipitation with GARS antibodies to compare interaction partners between wild-type and mutant GARS

    • Investigation of altered binding to tRNA synthetase complex components

    • Identification of novel pathological interactions specific to disease models

  • Histopathological Examination:

    • Immunohistochemistry with GARS antibodies on patient-derived tissues or animal models

    • Comparative analysis of GARS distribution patterns in healthy versus diseased neurons

    • Detection of abnormal GARS aggregation or inclusion bodies in affected tissues

  • Mechanism Investigation:

    • Tracking of non-canonical functions of GARS that might be specifically altered in disease states

    • Analysis of enzymatic activity changes using activity assays after immunoprecipitation

    • Investigation of potential gain-of-function mechanisms in neurodegenerative conditions

For these applications, antibodies must be carefully validated to ensure they recognize both wild-type and mutant forms of GARS, unless mutation-specific antibodies are being developed. The choice of fixation and antigen retrieval methods is particularly important for neuronal tissues, with TE buffer pH 9.0 being recommended for certain GARS antibodies in IHC applications .

How can researchers troubleshoot cross-reactivity issues with GARS antibodies?

Cross-reactivity presents a significant challenge when working with GARS antibodies. The following methodological approaches can help identify and address these issues:

  • Identifying Cross-Reactivity Problems:

    • Multiple unexpected bands in Western blot

    • Non-specific staining patterns in IHC/IF that don't match known GARS distribution

    • Positive signals in GARS-negative controls

    • Detection of proteins at molecular weights different from GARS (75-80 kDa)

    • Inconsistent results between different detection methods

  • Systematic Troubleshooting Approaches:

    a) Antibody Selection Refinement:

    • Switch to antibodies targeting unique regions of GARS with minimal homology to other proteins

    • Consider monoclonal antibodies for higher specificity when cross-reactivity is observed

    • Review comprehensive validation data for evidence of clean results in your specific application

    • Prioritize antibodies validated in knockout/knockdown systems

    b) Protocol Optimization:

    • Increase blocking stringency (longer time, different blocking agents like 5% BSA or commercial blockers)

    • Perform antibody titration experiments (test 4-5 dilutions spanning the recommended range)

    • Adjust incubation conditions (time, temperature, buffer composition)

    • Implement more stringent washing procedures (additional washes, higher detergent concentration)

    c) Application-Specific Solutions:

    For Western Blot:

    • Titrate primary antibody concentration (1:500-1:2000 dilution range)

    • Test alternative blocking agents (BSA vs. milk vs. commercial blockers)

    • Employ gradient gels for better separation of similar molecular weight proteins

    • Consider membrane stripping and sequential probing with different antibodies

    For Immunohistochemistry:

    • Optimize antibody dilution (1:50-1:500 recommended range)

    • Evaluate multiple antigen retrieval methods (TE buffer pH 9.0 vs. citrate buffer pH 6.0)

    • Include absorption controls with recombinant GARS protein

    • Compare with mRNA localization data when available

  • Confirmation Strategies:

    • Validate findings with orthogonal techniques when possible

    • Confirm critical results with genetic approaches (CRISPR knockout or siRNA knockdown)

    • Consider mass spectrometry to identify specific cross-reacting proteins

    • Document all optimization steps systematically for reproducibility

By methodically applying these troubleshooting approaches, researchers can minimize cross-reactivity issues and obtain more reliable, reproducible results with GARS antibodies across experimental systems.

How do GARS antibodies facilitate studying protein interactions within the multi-tRNA synthetase complex?

GARS antibodies serve as powerful tools for investigating protein-protein interactions within the multi-tRNA synthetase complex (MSC), providing insights into both structural organization and functional dynamics:

  • Complex Capture and Analysis:

    • GARS antibodies can immunoprecipitate the entire MSC from mammalian cell lysates

    • Research demonstrates that antibodies targeting individual MSC members can successfully co-precipitate all complex components across multiple cell types

    • This approach enables comprehensive identification of both direct and indirect interaction partners

  • Interaction Mapping Approaches:

    • Co-immunoprecipitation with GARS antibodies followed by mass spectrometry identifies the complete interaction network

    • Proximity-based assays using GARS antibodies (proximity ligation, BioID) reveal spatial organization

    • Crosslinking immunoprecipitation approaches capture transient or weak interactions

  • Complex Assembly Dynamics:

    • Time-course immunoprecipitation with GARS antibodies tracks temporal changes in complex composition

    • Analysis under various cellular conditions (stress, disease mutations) reveals context-dependent interactions

    • Size exclusion chromatography followed by GARS antibody detection identifies subcomplexes

  • Structural Analysis Applications:

    • Domain-specific GARS antibodies help map interaction interfaces within the complex

    • Epitope masking approaches identify specific binding regions

    • Conformation-specific antibodies can distinguish between free and complex-bound GARS states

  • Functional Impact Assessment:

    • GARS antibodies can selectively deplete specific GARS-containing complexes for functional studies

    • Comparison of aminoacylation activity in GARS-containing versus GARS-depleted complexes

    • Investigation of non-canonical functions associated with specific complex compositions

The recent development of recombinant high-affinity antibodies for aminoacyl-tRNA synthetases, including GARS, has significantly enhanced capabilities for studying these interactions . These validated antibodies provide reliable tools for capturing and analyzing the MSC, leading to improved understanding of both canonical and non-canonical functions.

What are the recommended storage and handling protocols for GARS antibodies?

Proper storage and handling of GARS antibodies is essential for maintaining performance and extending usable lifespan. The following protocols are recommended based on manufacturer guidelines:

  • Temperature Requirements:

    • Store at -20°C for long-term storage

    • For short-term use (up to 1 week), 2-8°C storage is acceptable

    • Avoid repeated freeze-thaw cycles that can cause protein denaturation

  • Aliquoting Recommendations:

    • Prepare small working aliquots (10-20 μl) to minimize freeze-thaw cycles

    • For some antibodies (like 15831-1-AP), manufacturers note that "Aliquoting is unnecessary for -20°C storage" for small sizes (20μl)

    • Use sterile microcentrifuge tubes and clear labeling for each aliquot

  • Buffer Composition Considerations:

    • Typical storage buffers include PBS with 0.02% sodium azide and 50% glycerol at pH 7.3

    • Some antibody preparations contain 0.1% BSA as a stabilizing agent

    • Avoid buffer substitutions unless specifically recommended by the manufacturer

  • Handling Best Practices:

    • Allow antibody to equilibrate to room temperature before opening to prevent condensation

    • Centrifuge briefly before opening to collect all liquid

    • Use clean pipette tips to prevent contamination

    • Wear gloves to prevent protein degradation from skin oils and proteases

    • Return to appropriate storage conditions promptly after use

  • Working Solution Preparation:

    • Prepare fresh working dilutions on the day of use whenever possible

    • Working solutions typically remain stable for up to 24 hours at 4°C

    • Extended storage of diluted antibodies is not recommended for critical applications

  • Stability Monitoring:

    • Properly stored antibodies typically remain stable for at least one year after shipment

    • Document performance over time to track potential degradation

    • Consider including a consistent positive control in experiments to monitor antibody performance

Following these storage and handling protocols will help ensure reproducible results across experiments and maximize the functional lifespan of GARS antibodies in research applications.

How does one determine the optimal GARS antibody for a specific experimental model?

Determining the optimal GARS antibody for a specific experimental model requires systematic evaluation following these methodological steps:

  • Initial Validation Data Assessment:

    • Review manufacturer's validation data for your specific model system

    • For example, 15831-1-AP has been validated in human brain tissue, HeLa cells, HepG2 cells, and HT-1080 cells for Western blot

    • IHC validation exists for human skin cancer tissue and mouse brain tissue

    • IF/ICC has been validated in HepG2 cells

  • Literature Precedent Analysis:

    • Search for published studies using GARS antibodies in your experimental model

    • Contact authors directly for specific protocols if needed

    • Review citations provided by antibody manufacturers for application-specific guidance

  • Species Reactivity Verification:

    • Confirm the antibody's species reactivity matches your experimental model

    • Common reactivity includes human, mouse, and rat for many GARS antibodies

    • For less common species, examine sequence homology in the epitope region

  • Preliminary Validation Experiments:

    Validation TechniqueKey ParametersControls to Include
    Western BlotSingle band at 75-80 kDaPositive control tissue/cells
    ImmunofluorescenceExpected subcellular patternSecondary antibody-only control
    ImmunohistochemistryAppropriate tissue distributionIsotype control
    ImmunoprecipitationSpecific pull-down of GARSIgG control
  • Optimization for Specific Sample Types:

    For Fixed Tissues:

    • Test recommended antigen retrieval methods (TE buffer pH 9.0 or citrate buffer pH 6.0)

    • Optimize antibody concentration based on signal-to-noise ratio

    • Evaluate different fixation protocols if possible

    For Cell Lines:

    • Consider different fixation/permeabilization methods

    • Optimize confluency levels to account for expression variation

    • Test both adherent and suspension preparation protocols if applicable

  • Comparative Analysis:

    • If resources permit, test multiple GARS antibodies in parallel

    • Evaluate based on signal-to-noise ratio, specificity, and reproducibility

    • Document optimization conditions systematically for future reference

By following this systematic approach, researchers can confidently determine the optimal GARS antibody for their specific experimental model and application requirements, ensuring reliable and reproducible results.

Product Science Overview

Introduction

Glycyl-tRNA synthetase (GARS) is an enzyme that plays a crucial role in protein synthesis by charging tRNA molecules with their corresponding amino acids, specifically glycine. This enzyme is part of the aminoacyl-tRNA synthetase family, which is essential for translating genetic information into functional proteins. GARS is encoded by the GARS1 gene in humans .

Structure and Function

GARS is a class II aminoacyl-tRNA synthetase that functions as an (alpha)2 dimer. It is responsible for ligating glycine to its corresponding tRNA (tRNA^Gly), a critical step in the translation process. This enzyme ensures that the genetic code is accurately translated into proteins by attaching the correct amino acid to its corresponding tRNA .

Clinical Significance

Mutations in the GARS1 gene have been implicated in several neurodegenerative diseases, including Charcot–Marie–Tooth disease type 2D (CMT2D) and distal spinal muscular atrophy type V (dSMA-V). These diseases are characterized by the degeneration of peripheral motor and sensory axons, leading to muscle weakness and atrophy, particularly in the distal extremities .

Mechanism of Disease

Research has shown that mutations in GARS can lead to toxic gain-of-function effects, which interfere with the normal function of the enzyme. These mutations do not necessarily disrupt the overall aminoacylation activity but can cause ribosome stalling and activation of the integrated stress response. This results in translational repression, affecting both the elongation and initiation stages of protein synthesis .

Autoantibodies and Immune Response

Interestingly, GARS has also been identified as a target of autoantibodies in certain autoimmune diseases, such as polymyositis and dermatomyositis. These autoantibodies can interfere with the normal function of GARS, contributing to the pathology of these diseases .

Research and Therapeutic Potential

Ongoing research is focused on understanding the precise mechanisms by which GARS mutations lead to disease and exploring potential therapeutic interventions. For example, studies have investigated the role of GARS in maintaining peripheral axons and its involvement in the integrated stress response .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.