JEM1 Antibody

Shipped with Ice Packs
In Stock

Description

JEM1 Protein Overview

JEM1 is a DnaJ-like chaperone localized to the ER membrane. It facilitates nuclear fusion (karyogamy) during yeast mating by interacting with Kar2p (yeast BiP) and Kar5p . Key features include:

PropertyDetail
Gene NameKAR8/JEM1 (YJL073W in S. cerevisiae)
Protein StructurePeripheral ER membrane protein with a J domain (47% identity to E. coli DnaJ)
Molecular FunctionChaperone activity, nuclear membrane fusion, ER stress response
Cellular ProcessRequired for outer and inner nuclear membrane fusion during mating

Role in Nuclear Fusion

  • JEM1 interacts with Kar2p (BiP) and Kar5p to form a fusogenic complex near the spindle pole body (SPB) .

  • Mutants (jem1Δ) exhibit bilateral karyogamy defects, where nuclei congress but fail to fuse .

  • Overexpression of JEM1 suppresses mating defects in kar2-1 mutants, indicating functional synergy with Kar2p .

Genetic and Biochemical Interactions

  • Genetic Interactions:

    • Synthetic lethality with kar5 and kar7/sec71 mutants .

    • Required for the stability of Kar5p, a protein critical for SPB fusion .

  • Biochemical Interactions:

    • Affinity capture-MS studies confirm JEM1’s interaction with ER-resident proteins, including Sec63p and Sec72p .

Applications of JEM1 Antibody

JEM1 antibody has been utilized in diverse experimental approaches:

ApplicationUtility
ImmunofluorescenceLocalizes JEM1 to the ER membrane and SPB in yeast zygotes .
Western BlotDetects JEM1 expression under ER stress conditions or during mating .
Genetic StudiesValidates JEM1 knockout strains and screens for interacting partners .

Research Implications

  • Model for Human Diseases: JEM1 homologs in mammals (e.g., ERdj5) are implicated in ER-associated degradation (ERAD) and neurodegenerative diseases .

  • Therapeutic Insights: Understanding JEM1’s role in membrane fusion could inform therapies targeting viral entry or intracellular pathogen resistance .

Future Directions

  • Characterize JEM1’s role in ER stress responses beyond mating.

  • Explore cross-species functional conservation using JEM1 antibody in mammalian models.

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
JEM1 antibody; KAR8 antibody; YJL073W antibody; HRC558 antibody; J1083 antibody; DnaJ-like chaperone JEM1 antibody; DnaJ-like protein of the ER membrane 1 antibody
Target Names
JEM1
Uniprot No.

Target Background

Function
This antibody targets JEM1, a DnaJ-like chaperone essential for nuclear membrane fusion during mating. JEM1 plays a crucial role in the process of cell fusion.
Gene References Into Functions
  1. Jem1p facilitates nuclear inner-membrane fusion and spindle pole body (SPB) fusion. PMID: 19090812
Database Links

KEGG: sce:YJL073W

STRING: 4932.YJL073W

Subcellular Location
Endoplasmic reticulum membrane; Single-pass type IV membrane protein. Nucleus membrane.

Q&A

What is JEM1 and why is it important for research?

JEM1 is a reported synonym of the BLZF1 gene, which encodes basic leucine zipper nuclear factor 1. This protein is known to function in Golgi organization, making it important for studying cellular trafficking and organelle structure. The human version of JEM1 has a canonical amino acid length of 400 residues and a protein mass of 44.9 kilodaltons, with two identified isoforms. It localizes primarily to the nucleus and Golgi apparatus and is widely expressed across multiple tissue types. Other names for this target include GOLGIN-45 and JEM-1 . Understanding JEM1's function contributes to our knowledge of cellular architecture and protein transport mechanisms.

What are the most common applications for JEM1 antibodies?

JEM1 antibodies are primarily used in several key applications including Western Blot (WB), Enzyme-Linked Immunosorbent Assay (ELISA), Immunofluorescence (IF), and Immunohistochemistry (IHC) . These techniques allow researchers to detect, quantify, and visualize the JEM1 protein in various experimental contexts. Western Blotting enables protein detection and approximate molecular weight determination, while ELISA provides quantitative measurements. Immunofluorescence and immunohistochemistry allow visualization of cellular localization and tissue distribution patterns, confirming JEM1's reported nuclear and Golgi localization.

How should I select the appropriate JEM1 antibody for my experiment?

When selecting a JEM1 antibody, consider the following methodological approach:

  • Determine your experimental application: Different applications require antibodies with different properties. For instance, Western blot applications may tolerate partially denatured epitopes, while immunofluorescence often requires antibodies recognizing native conformations .

  • Consider species reactivity: Available JEM1 antibodies have varying reactivity profiles, including those specific for bacteria (Biorbyt) or Saccharomyces (CUSABIO and MyBioSource) . Select an antibody with confirmed reactivity against your species of interest.

  • Evaluate validation data: Due to the "antibody characterization crisis," it's crucial to assess the quality of validation data provided by vendors. Look for evidence of specificity testing using knockout controls, immunoprecipitation followed by mass spectrometry, or other rigorous validation methods .

  • Check conjugation status: Determine whether your application requires a conjugated or unconjugated antibody. Commercially available JEM1 antibodies include unconjugated variants suitable for flexible detection strategies .

  • Review literature: Examine published research using JEM1 antibodies to identify reliably performing products and established protocols.

What controls should I implement when using JEM1 antibodies?

Implementing appropriate controls is essential for generating reliable and reproducible results with JEM1 antibodies. A methodological approach to controls should include:

  • Negative controls:

    • Isotype controls using irrelevant antibodies of the same isotype and concentration as your JEM1 antibody

    • Cell or tissue samples lacking JEM1 expression (if available)

    • Secondary antibody-only controls to assess non-specific binding

    • Blocking peptide competition assays to confirm specificity

  • Positive controls:

    • Cell lines or tissues with confirmed JEM1/BLZF1 expression

    • Recombinant JEM1 protein for Western blot or ELISA applications

    • Transfected cells overexpressing tagged JEM1 protein

  • Validation controls:

    • Knockdown/knockout validation where JEM1 expression is reduced/eliminated

    • Multiple antibodies against different epitopes of JEM1

    • Correlation between protein and mRNA levels

Implementing these controls addresses the antibody reproducibility concerns highlighted in recent literature and ensures confidence in experimental outcomes .

What are optimal conditions for Western blot detection of JEM1?

For optimal Western blot detection of JEM1 (BLZF1), follow this methodological approach:

  • Sample preparation:

    • Extract proteins using a buffer containing protease inhibitors to prevent degradation

    • For nuclear and Golgi proteins like JEM1, consider subcellular fractionation to enrich target proteins

    • Use appropriate detergents (e.g., 0.1% SDS, 1% Triton X-100) for solubilization

  • Gel electrophoresis:

    • Use 10-12% polyacrylamide gels suitable for the ~45 kDa molecular weight of JEM1

    • Load 20-50 μg of total protein per lane depending on expression levels

  • Transfer conditions:

    • Wet transfer at 100V for 1 hour or 30V overnight at 4°C

    • Use PVDF membrane for better protein retention

  • Blocking and antibody incubation:

    • Block with 5% non-fat dry milk or BSA in TBS-T for 1 hour at room temperature

    • Incubate with primary JEM1 antibody (1:500-1:2000 dilution) overnight at 4°C

    • Wash 3-5 times with TBS-T

    • Incubate with appropriate secondary antibody (1:5000-1:10000) for 1 hour at room temperature

  • Detection:

    • Use enhanced chemiluminescence (ECL) or fluorescent secondary antibodies

    • Expected band: approximately 44.9 kDa for canonical JEM1 protein

  • Verification:

    • Confirm specificity using blocking peptides

    • Consider double-checking with antibodies from different suppliers or clones

How can I optimize immunofluorescence staining with JEM1 antibodies?

Optimizing immunofluorescence staining with JEM1 antibodies requires careful attention to fixation, permeabilization, and antibody incubation conditions:

  • Sample preparation:

    • Fixation: For Golgi proteins like JEM1, use 4% paraformaldehyde for 15-20 minutes at room temperature

    • Permeabilization: Use 0.1-0.5% Triton X-100 for 5-10 minutes for nuclear proteins; alternatively, methanol fixation/permeabilization may preserve Golgi structure better

    • Blocking: Incubate with 5-10% normal serum (matching secondary antibody species) for 1 hour

  • Antibody incubation:

    • Primary antibody: Dilute JEM1 antibody 1:100-1:500 in blocking buffer

    • Incubation time: Overnight at 4°C or 1-2 hours at room temperature

    • Washing: Wash 3-5 times with PBS between antibody incubations

  • Detection and visualization:

    • Use fluorophore-conjugated secondary antibodies (1:200-1:1000)

    • Include nuclear counterstain (DAPI or Hoechst)

    • For co-localization studies, include Golgi markers (GM130, TGN46) to confirm JEM1 localization

  • Controls:

    • Include peptide competition controls

    • Use secondary-only controls

    • Consider co-staining with known Golgi markers to validate localization

  • Imaging parameters:

    • Use confocal microscopy for precise localization

    • Adjust exposure settings to avoid saturation

    • Capture multiple Z-sections for thorough examination of subcellular localization

How can I verify the specificity of my JEM1 antibody?

Verifying antibody specificity is crucial given the antibody characterization crisis highlighted in recent literature . A methodological approach to verifying JEM1 antibody specificity includes:

  • Genetic approaches:

    • Test antibody in JEM1/BLZF1 knockout or knockdown systems

    • Compare staining patterns in cells with varying JEM1 expression levels

    • Use siRNA or CRISPR to reduce target expression and confirm signal reduction

  • Biochemical validation:

    • Perform peptide competition assays where the antibody is pre-incubated with purified JEM1 protein

    • Run immunoprecipitation followed by mass spectrometry to confirm target identity

    • Test multiple antibodies against different epitopes of JEM1

  • Orthogonal methods:

    • Compare protein detection with mRNA levels using qPCR

    • Use GFP-tagged JEM1 constructs to compare with antibody staining patterns

    • Employ proximity ligation assays with two different JEM1 antibodies

  • Cross-reactivity assessment:

    • Test the antibody against closely related proteins

    • Evaluate species cross-reactivity if working with non-human models

    • Check for unexpected bands in Western blots or staining patterns in immunofluorescence

This multi-faceted approach addresses the concerns about antibody specificity that have been identified as a major source of irreproducibility in biomedical research .

What are the key considerations when interpreting contradictory results with JEM1 antibodies?

When faced with contradictory results using JEM1 antibodies, apply this systematic troubleshooting approach:

  • Antibody-related factors:

    • Epitope specificity: Different antibodies may recognize different epitopes or isoforms of JEM1

    • Clone variation: Compare monoclonal vs. polyclonal antibodies and results from different vendors

    • Lot-to-lot variability: Check if different lots of the same antibody produce consistent results

    • Validation data: Review comprehensive validation data provided by vendors or published literature

  • Experimental factors:

    • Sample preparation: Different fixation and permeabilization methods may affect epitope accessibility

    • Antibody concentration: Titrate antibodies to determine optimal working concentration

    • Incubation conditions: Vary temperature, time, and buffer compositions

    • Detection methods: Compare different visualization techniques

  • Biological factors:

    • Isoform expression: Consider whether tissue-specific isoforms might explain differences

    • Post-translational modifications: Determine if modifications might mask epitopes in certain conditions

    • Protein interactions: Assess whether protein-protein interactions might block antibody binding

    • Expression levels: Evaluate whether expression levels vary across experimental conditions

  • Resolution strategies:

    • Use multiple antibodies targeting different epitopes

    • Implement orthogonal detection methods

    • Consider advanced techniques like proximity ligation assays

    • Consult with antibody manufacturers for technical support

How do I address batch-to-batch variability in JEM1 antibodies?

Batch-to-batch variability is a significant challenge in antibody-based research. To address this issue with JEM1 antibodies:

  • Proactive measures:

    • Purchase larger quantities of a single, well-performing lot when possible

    • Document lot numbers and retain small aliquots of previous lots for comparison

    • Request certificate of analysis and validation data for each new lot

    • Consider using recombinant antibodies which typically show less lot-to-lot variation

  • Validation protocols for new batches:

    • Perform side-by-side comparisons with previous lots

    • Establish a panel of positive and negative control samples

    • Create standard curves for quantitative applications

    • Document staining patterns and signal intensities for qualitative comparison

  • Standardization approaches:

    • Normalize results using internal controls

    • Include standard samples across experiments

    • Develop and maintain detailed standard operating procedures

    • Consider collaborative testing with other laboratories

  • Documentation and reporting:

    • Record lot numbers in all experimental notes and publications

    • Report batch effects and variations in research papers

    • Contact manufacturers about significant variations between lots

    • Share experiences through antibody validation databases

How can I use JEM1 antibodies for co-immunoprecipitation and protein interaction studies?

To use JEM1 antibodies effectively for co-immunoprecipitation (co-IP) and protein interaction studies:

  • Buffer optimization:

    • Use mild lysis buffers (e.g., 1% NP-40 or 0.5% Triton X-100) to preserve protein-protein interactions

    • Include protease and phosphatase inhibitors to maintain protein integrity

    • Consider crosslinking agents for transient or weak interactions

    • Test different salt concentrations (150-300 mM NaCl) to optimize specificity

  • Antibody selection and coupling:

    • Choose antibodies with demonstrated specificity for JEM1/BLZF1

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

    • Consider directly conjugating antibodies to beads to avoid co-elution of antibody heavy chains

    • Use controls including isotype-matched irrelevant antibodies

  • Methodological approach:

    • Prepare cell lysates under non-denaturing conditions

    • Pre-clear with Protein A/G beads for 1 hour at 4°C

    • Incubate cleared lysates with JEM1 antibody overnight at 4°C

    • Add fresh Protein A/G beads and incubate for 2-4 hours

    • Wash extensively with lysis buffer (4-5 times)

    • Elute bound proteins with SDS sample buffer or specific peptides

  • Analysis and verification:

    • Analyze by Western blot for specific interacting proteins

    • Consider mass spectrometry for unbiased identification of binding partners

    • Confirm interactions using reverse co-IPs

    • Validate physiological relevance through functional assays

What are common pitfalls when using JEM1 antibodies and how can they be avoided?

Common pitfalls when using JEM1 antibodies and strategies to avoid them include:

  • Non-specific binding:

    • Pitfall: High background or multiple bands in Western blot

    • Solution: Increase blocking time/concentration, optimize antibody dilution, include additional washing steps, use more stringent washing buffers, consider alternative blocking agents

  • Loss of signal over time:

    • Pitfall: Decreased antibody performance with storage

    • Solution: Store antibodies according to manufacturer recommendations, aliquot to avoid freeze-thaw cycles, add preservatives like sodium azide, monitor storage conditions

  • Inconsistent results:

    • Pitfall: Variable staining patterns or band intensities

    • Solution: Standardize protocols, use positive controls, document lot numbers, implement quality control measures

  • Poor reproducibility:

    • Pitfall: Inability to reproduce published results

    • Solution: Request detailed protocols from authors, contact antibody manufacturers for technical support, systematically optimize conditions

  • Epitope masking:

    • Pitfall: Loss of signal due to post-translational modifications or protein interactions

    • Solution: Try multiple antibodies against different epitopes, test different sample preparation methods, consider native vs. denaturing conditions

  • Cross-reactivity:

    • Pitfall: Antibody recognizing proteins other than JEM1

    • Solution: Validate specificity using knockout controls, peptide competition, and orthogonal methods

  • Fixation artifacts:

    • Pitfall: Altered staining patterns due to fixation

    • Solution: Compare multiple fixation methods, optimize fixation time and conditions

How can I quantitatively measure JEM1 protein levels in complex samples?

For quantitative measurement of JEM1 protein levels in complex samples, consider these methodological approaches:

  • Quantitative Western blotting:

    • Use a standard curve of recombinant JEM1 protein

    • Include loading controls (GAPDH, β-actin, or total protein stains)

    • Employ fluorescent secondary antibodies for wider linear range

    • Analyze using software like ImageJ or commercial alternatives

    • Normalize to appropriate controls

  • ELISA-based approaches:

    • Develop sandwich ELISA using two antibodies recognizing different JEM1 epitopes

    • Generate standard curves using purified JEM1 protein

    • Validate assay specificity and sensitivity

    • Determine limits of detection and quantification

    • Consider commercial ELISA kits if available

  • Mass spectrometry-based quantification:

    • Use targeted approaches like selected reaction monitoring (SRM)

    • Develop signature peptides unique to JEM1

    • Include isotopically labeled standards for absolute quantification

    • Validate method using samples with known JEM1 concentrations

  • Flow cytometry:

    • Optimize cell permeabilization for intracellular JEM1 detection

    • Use fluorophore-conjugated JEM1 antibodies or primary-secondary combinations

    • Include isotype controls and calibration beads

    • Analyze median fluorescence intensity for relative quantification

MethodSensitivityThroughputAdvantagesLimitations
Western BlotMediumLowSize information, widely accessibleSemi-quantitative, time-consuming
ELISAHighHighHigh specificity, quantitativeNo size information, requires two specific antibodies
Mass SpectrometryHighMediumHighly specific, can be multiplexedExpensive equipment, complex analysis
Flow CytometryMedium-HighHighSingle-cell resolution, multi-parameterRequires cell suspensions, complex optimization

How can I use JEM1 antibodies in super-resolution microscopy?

Adapting JEM1 antibodies for super-resolution microscopy requires specific optimization strategies:

  • Antibody preparation:

    • Use highly specific primary antibodies with validated specificity

    • Consider directly conjugated primary antibodies to reduce steric hindrance

    • For secondary antibody approaches, use F(ab)2 fragments for smaller size

    • Optimize antibody concentration to achieve appropriate labeling density

  • Sample preparation:

    • Use thin sections (70-100 nm) for techniques like STORM or PALM

    • Optimize fixation to preserve ultrastructure while maintaining epitope accessibility

    • Consider using specialized fixatives like glyoxal for improved structure preservation

    • For Golgi proteins like JEM1, gentle fixation and permeabilization are crucial

  • Technique-specific considerations:

    • STORM/PALM: Use photoconvertible fluorophores with appropriate buffer systems

    • STED: Select fluorophores with appropriate photostability

    • SIM: Ensure even staining and minimize background

    • Expansion microscopy: Test antibody retention after expansion

  • Controls and validation:

    • Include co-localization with known Golgi markers

    • Compare with conventional confocal microscopy

    • Use multiple antibodies against different JEM1 epitopes

    • Implement appropriate negative controls

  • Analysis and interpretation:

    • Apply suitable reconstruction algorithms

    • Perform quantitative analysis of localization patterns

    • Consider using clustering algorithms to analyze protein distribution

    • Validate findings with orthogonal approaches

What are the latest advances in JEM1 antibody development and characterization?

Recent advances in antibody development and characterization relevant to JEM1 antibodies include:

  • Recombinant antibody technology:

    • Development of recombinant antibodies with defined sequences

    • Improved consistency and reduced batch-to-batch variation

    • Enhanced reproducibility addressing the "antibody characterization crisis"

    • Potential for engineering improved specificity and affinity

  • Advanced validation methods:

    • Implementation of knockout validation using CRISPR/Cas9

    • Independent epitope approach using multiple antibodies

    • Orthogonal validation comparing protein and mRNA levels

    • Mass spectrometry confirmation of immunoprecipitated targets

  • Novel antibody formats:

    • Development of nanobodies and single-domain antibodies

    • Bispecific antibodies for enhanced specificity

    • Intrabodies for live-cell applications

    • Site-specific conjugation methods for improved performance

  • Reporting standards and databases:

    • Implementation of antibody validation initiative guidelines

    • Development of antibody validation databases

    • Improved reporting standards in publications

    • Community efforts to share validation data across laboratories

  • Application-specific optimization:

    • Customized antibodies for super-resolution microscopy

    • Development of antibodies suitable for live-cell imaging

    • Expansion of species reactivity for comparative studies

    • Enhanced sensitivity for detecting low-abundance proteins

How can I use computational approaches to predict potential cross-reactivity of JEM1 antibodies?

Computational approaches can help predict potential cross-reactivity of JEM1 antibodies, enhancing experimental design and troubleshooting:

  • Epitope analysis:

    • Identify the JEM1 epitope recognized by your antibody

    • Perform BLAST searches to identify proteins with similar sequences

    • Use epitope prediction algorithms to assess potential cross-reactivity

    • Consider three-dimensional structural similarity beyond sequence homology

  • Homology assessment:

    • Compare JEM1 sequence with related proteins in the basic leucine zipper family

    • Identify conserved domains that might lead to cross-reactivity

    • Consider species conservation when working across model organisms

    • Evaluate potential post-translational modification sites

  • Machine learning approaches:

    • Apply machine learning algorithms trained on antibody-epitope interactions

    • Use existing antibody cross-reactivity datasets to inform predictions

    • Implement ensemble methods combining multiple prediction approaches

    • Validate computational predictions experimentally

  • Structural biology integration:

    • Use protein structure databases to assess epitope accessibility

    • Perform molecular docking simulations between antibody and potential targets

    • Consider conformational epitopes that may not be evident from sequence alone

    • Evaluate impact of protein dynamics on epitope recognition

  • Experimental validation of predictions:

    • Test antibody against predicted cross-reactive proteins

    • Perform competitive binding assays with potential cross-reactive peptides

    • Use arrays of peptides or proteins to systematically assess cross-reactivity

    • Correlate computational predictions with experimental outcomes

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.