MROH8 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to MROH8 Antibody

The MROH8 antibody is a highly specific immunoglobulin designed to target the MROH8 protein, a member of the maestro heat-like repeat family. While the exact biological function of MROH8 remains under investigation, recent studies have implicated it in cellular processes such as transcriptional regulation and tumor suppression, particularly in pancreatic cancer .

Antibody Structure and Mechanism

Antibody Class: The MROH8 antibody is classified as a monoclonal IgG (immunoglobulin G), optimized for high-affinity binding to epitopes on the MROH8 protein.
Binding Regions:

  • Fab Fragments: Contain variable domains (V_H and V_L) that interact with MROH8’s conserved regions.

  • Fc Region: Facilitates interactions with effector molecules, enabling applications like immunoprecipitation or Western blotting .

Role in Pancreatic Cancer

MROH8 has been identified as a tumor suppressor in pancreatic cancer, with its expression inversely correlated with tumor metastasis. Antibody-based assays (e.g., Western blotting) revealed that:

  • MROH8 Overexpression: Inhibits tumor cell growth and metastasis by downregulating CAPN2 (calpain-2), a pro-metastatic protease .

  • MROH8 Knockdown: Significantly enhances tumor colony formation (P<0.001) .

Regulatory Pathway

The MROH8 antibody is central to studying its upstream and downstream interactions:

ComponentFunctionAntibody Utility
METTL16m6A writerDetects m6A-mediated stabilization of MROH8 mRNA .
YTHDC2m6A readerConfirms MROH8 mRNA degradation via YTHDC2 .
TBPTranscription factorBinds to CAPN2 promoter, modulated by MROH8 .

Clinical Implications

  • Survival Correlation: High MROH8 expression correlates with improved survival in pancreatic cancer patients (P<0.001) .

  • Therapeutic Target: The METTL16–MROH8–TBP pathway offers potential for targeted therapies, validated via antibody-mediated knockdown/knockout experiments .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery timelines, please consult your local distributor.
Synonyms
MROH8 antibody; C20orf131 antibody; C20orf132 antibody; Protein MROH8 antibody; Maestro heat-like repeat-containing protein family member 8 antibody
Target Names
MROH8
Uniprot No.

Q&A

What is MROH8 and what is known about its biological function?

MROH8 (Maestro Heat-like Repeat-containing Protein Family Member 8) is a protein encoded by the MROH8 gene located on chromosome 20. It is also known by several alternative names including C20orf131, C20orf132, dJ621N11.3, and dJ621N11.4 .

Recent research in pancreatic cancer has identified MROH8 as part of a regulatory pathway involving N6-methyladenosine (m6A) RNA modifications. MROH8 appears to function by modulating transcription factor activity, particularly through interaction with TATA-binding protein (TBP). Specifically, MROH8 has been shown to:

  • Negatively regulate TBP by promoting its degradation after binding

  • Function downstream of the m6A writer METTL16

  • Correlate with improved survival in pancreatic cancer patients

  • Inhibit tumor cell growth when overexpressed

This emerging evidence suggests MROH8 may act as a tumor suppressor through transcriptional regulation mechanisms, though more research is needed to fully elucidate its functions across different tissues and cellular contexts.

What types of MROH8 antibodies are available for research applications?

Antibody TypeHost SpeciesTarget RegionApplicationsReactivityExamples
PolyclonalRabbitFull proteinWB, IHC, ICC/IFHuman, MouseNovus Biologicals NBP2-34133
PolyclonalRabbit379-429 aaWBHuman, Rat, MouseSt. John's Labs STJ198035
PolyclonalRabbit1-275 AAELISA, WBHumanBiomatik CAC15880
PolyclonalRabbitMiddle regionWBHuman, MouseFisher Scientific NBP157772
PolyclonalMouseNot specifiedWB, IFHumanMyBioSource offerings
ConjugatedRabbitNot specifiedELISAHumanHRP/Biotin-conjugated variants

Most commercially available MROH8 antibodies are polyclonal, derived primarily from rabbits, with different epitope targets allowing researchers to select antibodies appropriate for their specific experimental needs .

What validation methods should I use to confirm MROH8 antibody specificity?

Proper validation of MROH8 antibodies is critical for ensuring experimental reliability. A comprehensive validation strategy should include:

  • Western blot molecular weight verification:

    • Confirm detection at the expected molecular weight for MROH8

    • Some antibodies detect bands of approximately 140-150 kDa, consistent with predicted MROH8 size

  • Positive and negative controls:

    • Use cell lines with known MROH8 expression levels

    • Implement MROH8 knockdown/knockout samples as negative controls

    • Research shows that MROH8 knockdown increases cell colony formation while overexpression inhibits tumor cell growth

  • Peptide competition assays:

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

    • For example, peptide sequences like "PHLENLDTIIKLPLRFQRLGHLVALMALLCGDPQEKVAEEAAEGIHSLLH" for NBP157772

  • Protein array testing:

    • Some manufacturers verify specificity on protein arrays containing target protein plus hundreds of non-specific proteins

  • Cross-application validation:

    • Confirm consistency across multiple applications (e.g., WB, IHC, IF)

    • Document optimal dilutions for each application (e.g., 1:500-1:1000 for IHC )

  • Correlation with mRNA expression data:

    • Compare protein detection with transcriptomic data

    • MROH8 mRNA stability has been shown to be enhanced by m6A modifications mediated by METTL16

Always document validation procedures thoroughly and include appropriate controls in all experiments to ensure reliable interpretation of results.

How should I design experiments to study MROH8's role in transcriptional regulation?

MROH8 has been identified as a regulator of transcription factor activity, particularly through its interaction with TBP. To investigate this function:

  • Protein-protein interaction studies:

    • Co-immunoprecipitation (Co-IP): Use MROH8 antibodies to pull down protein complexes and probe for transcription factors like TBP

    • Reciprocal Co-IP: Use TBP antibodies to confirm interaction from both directions

    • Research has demonstrated that MROH8 physically binds to TBP protein

  • Protein degradation analysis:

    • Cycloheximide chase assays: Monitor TBP degradation rates with/without MROH8 overexpression

    • Proteasome inhibitor studies: Determine if MROH8-mediated TBP degradation is proteasome-dependent

    • Temporal analysis: In published studies, inducing MROH8 overexpression and then withdrawing the inducer showed that decreasing MROH8 levels led to increasing TBP levels

  • Transcriptional impact assessment:

    • Luciferase reporter assays: Measure transcriptional activity of TBP-dependent promoters

    • ChIP-seq analysis: Assess genome-wide changes in TBP binding with MROH8 modulation

    • RNA-seq: Identify genes differentially expressed with MROH8 manipulation

  • Domain mapping experiments:

    • Generate MROH8 truncation mutants to identify regions critical for TBP interaction

    • Use antibodies targeting different MROH8 domains to potentially disrupt specific interactions

  • Functional validation:

    • MROH8 knockdown and overexpression studies in relevant cell lines

    • Rescue experiments with wild-type vs. interaction-deficient MROH8 mutants

    • Published data shows MROH8 knockdown promotes cell colony formation while overexpression inhibits tumor cell growth

A comprehensive experimental design should incorporate multiple methodological approaches to provide robust evidence for MROH8's transcriptional regulatory functions.

What experimental strategies can optimize detection of MROH8 in clinical samples with low expression?

Detecting low-abundance proteins like MROH8 in clinical samples requires specialized approaches:

  • Signal amplification methods:

    • Tyramide signal amplification (TSA): Can increase detection sensitivity 10-100 fold for IHC/IF

    • Polymer-based detection systems: Often provide better signal-to-noise ratio than ABC methods

    • Higher-sensitivity substrates: Super Signal or femto-level chemiluminescent reagents for Western blotting

  • Sample preparation optimization:

    ProcedureStandard MethodEnhanced Method for Low Expression
    Antigen retrievalCitrate buffer pH 6.0Test multiple buffers (citrate pH 6.0, EDTA pH 8.0, Tris-EDTA pH 9.0)
    Antibody incubation1 hour at RTOvernight at 4°C with optimized concentration
    BlockingStandard BSA or serumSpecialized blockers to reduce background
    Signal detectionStandard DABAmplified detection systems (TSA, QD)
  • Antibody selection and optimization:

    • Choose antibodies validated for high sensitivity (e.g., those confirmed for detecting endogenous levels)

    • Perform careful titration experiments to determine optimal concentration

    • For IHC/IF: 1:500-1:1000 dilutions are typically recommended

    • For Western blot: 1:1000-2000 or 1.0 μg/ml concentrations are often optimal

  • Pre-enrichment strategies:

    • Immunoprecipitation before Western blotting to concentrate MROH8

    • Laser capture microdissection to isolate specific cell populations

  • Alternative or complementary detection methods:

    • RNAscope for mRNA detection to complement protein analysis

    • Multiplexed detection to visualize MROH8 alongside known interacting partners

These strategies can significantly improve detection sensitivity for MROH8 in clinical samples, enabling more reliable analysis in experimental and diagnostic contexts.

How does MROH8 interact with the m6A modification pathway in cancer cells?

Recent research has revealed a novel regulatory axis involving MROH8 and m6A RNA modifications in pancreatic cancer:

  • METTL16-MROH8-TBP-CAPN2 regulatory axis:

    • METTL16 (an m6A writer) enhances MROH8 mRNA stability through m6A modifications

    • MROH8 negatively regulates TBP by promoting its degradation

    • TBP functions as a transcription factor for CAPN2, which promotes tumor growth

    • This pathway effectively links RNA modifications to transcriptional regulation and cancer progression

  • Experimental evidence:

    • METTL16 overexpression significantly increases MROH8 levels and decreases CAPN2 levels

    • METTL16 and YTHDC2 (an m6A reader) collaboratively enhance MROH8 mRNA stability

    • MROH8 negatively correlates with CAPN2 (r=-0.3, P<0.001)

    • MROH8 positively correlates with better survival outcomes in pancreatic cancer patients

  • Methodological approach to study this pathway:

    • m6A-specific quantitative PCR to measure modification levels

    • RNA immunoprecipitation using m6A antibodies followed by MROH8 detection

    • Co-IP experiments between MROH8 and transcription factors

    • Functional assays measuring tumor proliferation and metastasis with pathway manipulation

  • Experimental design considerations:

    • Use antibodies against multiple pathway components (METTL16, MROH8, TBP, CAPN2)

    • Implement both gain-of-function and loss-of-function approaches

    • Validate findings across cell lines, patient-derived organoids, and animal models

    • Consider temporal dynamics of the pathway activation

This regulatory mechanism represents an important area for further research, potentially offering new therapeutic targets for pancreatic and other cancers.

What are the optimal protocols for using MROH8 antibodies in multiplexed immunofluorescence studies?

Multiplexed immunofluorescence (mIF) allows simultaneous visualization of MROH8 alongside other proteins of interest. Here's a comprehensive protocol:

  • Antibody selection and panel design:

    • Choose MROH8 antibodies validated for IF applications

    • Consider host species compatibility with other antibodies in your panel

    • If using multiple rabbit antibodies, plan sequential staining with tyramide signal amplification

    • Example panel for studying MROH8 in cancer: MROH8 + TBP + CAPN2 + epithelial marker + immune markers

  • Step-by-step multiplexed staining protocol:

    a) Tissue preparation:

    • Use FFPE or frozen sections (4-6 μm thickness)

    • Perform deparaffinization and rehydration for FFPE samples

    • Conduct heat-induced epitope retrieval (optimal buffer determined empirically)

    b) Blocking and antibody application:

    • Block with 5-10% normal serum or specialized blocking buffer (30-60 minutes)

    • Apply primary antibodies sequentially or in compatible combinations

    • For MROH8: Use 1-4 μg/ml concentration or 0.25-2 μg/mL

    • Incubate overnight at 4°C for optimal sensitivity

    c) Detection system:

    • Use fluorophore-conjugated secondary antibodies or TSA systems

    • Include spectral unmixing controls if needed

    • If using sequential staining, include antibody stripping/quenching steps

  • Critical optimization parameters:

    ParameterRecommendationRationale
    Antibody dilution1:500-1:1000 (IHC)
    1-4 μg/ml (IF)
    Balance between signal and background
    Staining orderStart with MROH8 if low abundancePreserve epitope accessibility
    Antigen retrievalTest multiple buffersDifferent epitopes have different requirements
    Signal amplificationTSA for low-abundance targetsIncreases detection sensitivity
    ControlsSingle-color and FMO controlsEssential for accurate interpretation
  • Imaging and analysis considerations:

    • Use spectral imaging systems when possible

    • Employ consistent exposure settings across specimens

    • Utilize specialized software capable of cell segmentation and co-localization analysis

    • Quantify both expression levels and spatial relationships between proteins

Following these guidelines will enable successful integration of MROH8 detection into multiplexed immunofluorescence studies, providing valuable spatial context for understanding MROH8's functional relationships.

How can I optimize MROH8 antibody performance for protein-protein interaction studies?

Investigating MROH8's interactions with proteins like TBP requires optimized antibody-based techniques:

  • Co-immunoprecipitation (Co-IP) protocol optimization:

    a) Lysis buffer composition:

    • Base buffer: 50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA

    • Detergent: Use mild non-ionic detergents (0.5-1% NP-40 or 0.5% Triton X-100)

    • Protease/phosphatase inhibitors: Include complete protease inhibitor cocktail

    • Consider adding 10% glycerol for protein stability

    • DNase/RNase: Include if DNA/RNA bridging is a concern

    b) Antibody selection and preparation:

    • Choose MROH8 antibodies raised against regions unlikely to be involved in protein interactions

    • Pre-clear lysates with protein A/G beads before immunoprecipitation

    • Use 2-5 μg antibody per 500 μg total protein for optimal results

    • Consider pre-crosslinking antibody to beads to prevent antibody contamination in eluates

    c) Washing and elution conditions:

    • Use graduated stringency washes to reduce background

    • Optimize number of washes (typically 3-5) based on signal-to-noise ratio

    • Consider native elution with peptide competition when possible

  • Proximity Ligation Assay (PLA) optimization:

    • Requires antibodies from different host species against MROH8 and interaction partners

    • Optimal fixation: 4% paraformaldehyde for 15-20 minutes

    • Permeabilization: 0.2% Triton X-100 for 10 minutes

    • Primary antibody dilutions: Start with manufacturer's recommendations for IF and optimize

    • Include appropriate negative controls (omitting one primary antibody)

  • FRET/BRET approaches for live-cell interaction studies:

    • While not directly using antibodies, can complement antibody-based interaction studies

    • Useful for monitoring dynamic interactions in living cells

    • Can validate interactions detected by antibody-based methods

  • Validation strategies for interactions:

    • Reciprocal Co-IP (pull down with antibodies against both proteins)

    • Peptide competition to confirm specificity

    • IP-mass spectrometry to identify novel interaction partners

    • Published research confirms MROH8-TBP interaction through Co-IP experiments

These optimized protocols will enhance the reliability and sensitivity of protein-protein interaction studies involving MROH8, providing deeper insights into its functional roles.

What are the optimal storage and handling conditions for MROH8 antibodies?

Proper storage and handling of MROH8 antibodies is critical for maintaining their performance over time:

Storage ParameterRecommended ConditionsNotes
Short-term storage4°C (refrigeration)Typically stable for 1-2 weeks
Long-term storage-20°C in aliquotsAvoid repeated freeze-thaw cycles
Buffer formulationPBS with 40-50% glycerol, 0.02-0.09% sodium azideGlycerol prevents freezing damage
Additional stabilizersMay contain 0.5% BSA or 2% sucroseEnhances stability during storage
Aliquot size10-20 μL recommendedMinimizes freeze-thaw cycles
Thawing procedureGentle thawing at room temperature or 4°CAvoid rapid heating
Working dilution storageUp to 1 week at 4°CPrepare fresh dilutions for critical experiments

Most manufacturers recommend storing MROH8 antibodies at 4°C for short-term use and at -20°C for long-term storage . For lyophilized antibodies, manufacturers typically recommend reconstitution by centrifuging the vial at 12,000 x g for 20 seconds, adding the appropriate volume of distilled water, vortexing, and centrifuging again .

Always refer to the specific storage instructions provided by the manufacturer for your particular MROH8 antibody, as formulations may vary between suppliers.

How do different sample preparation methods affect MROH8 antibody performance in various applications?

Sample preparation significantly impacts MROH8 antibody performance across different applications:

  • Western Blotting sample preparation:

    ParameterStandard MethodOptimization for MROH8
    Lysis bufferRIPA bufferFor membrane proteins: Consider NP-40 or digitonin-based buffers
    Protein denaturation95°C for 5 minTest both boiled and non-boiled samples
    Reducing conditionsβ-mercaptoethanol or DTTEssential for accessing linear epitopes
    Loading amount10-30 μg total proteinMay need 30-50 μg for low abundance detection
    Transfer conditionsStandard wet transferConsider longer transfer times for high MW proteins

    Published MROH8 antibodies are validated for Western blot applications with specific recommended dilutions (1:1000-2000 or 1.0 μg/ml) .

  • Immunohistochemistry/Immunofluorescence sample preparation:

    ParameterEffect on MROH8 DetectionRecommendation
    Fixation methodAffects epitope accessibility10% neutral buffered formalin (standard FFPE)
    Fixation durationOverfixation can mask epitopes24-48 hours optimal for most tissues
    Antigen retrievalCritical for FFPE samplesHeat-induced epitope retrieval (HIER) in citrate buffer pH 6.0 or EDTA pH 9.0
    Section thicknessAffects signal intensity4-6 μm optimal for most applications
    Blocking solutionReduces background5-10% normal serum matching secondary antibody species

    MROH8 antibodies have been validated for IHC/IF applications with specific dilution recommendations (1:500-1:1000 for IHC, 1-4 μg/ml for IF) .

  • ELISA and other immunoassays:

    ParameterConsideration for MROH8Recommendation
    Coating bufferAffects protein binding to plateCarbonate-bicarbonate buffer pH 9.6
    Blocking agentPrevents non-specific binding1-5% BSA or 5% non-fat dry milk
    Sample dilutionAffects detection sensitivitySerial dilutions to determine optimal concentration
    Detection systemImpacts sensitivityHRP/TMB systems common; consider amplified detection for low abundance

    Some MROH8 antibodies are specifically validated for ELISA applications , including biotin-conjugated and HRP-conjugated variants for enhanced detection .

  • Flow cytometry considerations:

    • Fixation: 2-4% paraformaldehyde preferred for intracellular targets

    • Permeabilization: 0.1-0.5% saponin or 0.1% Triton X-100 for intracellular access

    • Blocking: Use 2-5% serum or BSA to reduce non-specific binding

    • Controls: Include appropriate isotype controls and FMO controls

By optimizing these sample preparation parameters for each application, researchers can maximize the performance of MROH8 antibodies and obtain more reliable and reproducible results.

What are the current limitations of MROH8 antibodies and how can these be addressed in research?

Despite their utility, current MROH8 antibodies have several limitations that researchers should address:

  • Specificity concerns:

    • Limitation: While some MROH8 antibodies have been tested on protein arrays , comprehensive specificity testing across diverse sample types may be limited.

    • Solution: Implement rigorous validation using multiple approaches:

      • Use positive and negative controls including MROH8 knockdown/knockout samples

      • Perform blocking peptide competition assays with immunizing peptides

      • Compare results across antibodies targeting different epitopes

  • Limited application validation:

    • Limitation: Many MROH8 antibodies are validated for specific applications only (e.g., WB or IHC) , restricting their utility in multi-modal studies.

    • Solution:

      • Conduct application-specific validation before use

      • Document and share validation results with the research community

      • Consider requesting additional validation data from manufacturers

  • Batch-to-batch variability:

    • Limitation: Particularly with polyclonal antibodies, batch-to-batch variability can affect experimental reproducibility.

    • Solution:

      • Purchase larger lots when possible

      • Include consistent positive controls across experiments

      • Consider switching to recombinant monoclonal antibodies when available, as these offer improved consistency

  • Detection sensitivity:

    • Limitation: MROH8 may be expressed at low levels in some tissues, challenging detection limits.

    • Solution:

      • Implement signal amplification techniques (TSA, enhanced chemiluminescence)

      • Optimize sample preparation to maximize epitope accessibility

      • Consider pre-enrichment strategies (IP before WB)

  • Cross-reactivity with related proteins:

    • Limitation: MROH8 belongs to the Maestro heat-like repeat family, which may share sequence homology with other members.

    • Solution:

      • Validate antibody specificity using recombinant proteins of related family members

      • Consider computational analysis of epitope sequences for potential cross-reactivity

      • Include appropriate controls when studying tissues expressing multiple family members

By acknowledging these limitations and implementing appropriate strategies to address them, researchers can maximize the reliability and reproducibility of their MROH8 research.

How can MROH8 antibodies be used in studying its role in cancer biology?

Recent research has identified MROH8 as a potential tumor suppressor with prognostic significance in pancreatic cancer . MROH8 antibodies can be used to investigate this role through several approaches:

  • Expression profiling across cancer types and stages:

    • Use validated MROH8 antibodies for IHC to assess expression in tumor microarrays

    • Compare expression between tumor tissue and adjacent normal tissue

    • Correlate expression levels with clinicopathological parameters

    • Research suggests MROH8 expression correlates with improved survival in pancreatic cancer

  • Mechanism investigation using cell line models:

    • Western blot analysis to confirm MROH8 expression changes after genetic manipulation

    • Co-IP studies to investigate protein interactions with transcription factors

    • ICC/IF to assess subcellular localization under different conditions

    • Published research shows MROH8 knockdown promotes cell colony formation while overexpression inhibits tumor cell growth

  • Molecular pathway analysis:

    • Study the METTL16-MROH8-TBP-CAPN2 axis identified in pancreatic cancer

    • Assess m6A modification impact on MROH8 expression

    • Investigate downstream effects on cell proliferation and metastasis

    • Use multiplexed approaches to simultaneously visualize multiple pathway components

  • Clinical correlation studies:

    • Develop standardized IHC scoring systems for MROH8 expression

    • Correlate expression with patient survival and treatment response

    • Consider MROH8 as a potential prognostic biomarker

    • Existing research shows association between MROH8 expression and improved prognosis in pancreatic cancer patients

  • Preclinical therapeutic targeting:

    • Study changes in MROH8 expression in response to therapies

    • Consider approaches to upregulate MROH8 as a potential therapeutic strategy

    • Investigate combination approaches targeting the MROH8 pathway

MROH8 antibodies provide essential tools for these investigations, enabling visualization and quantification of protein expression, localization, and interactions in cancer research contexts.

How can MROH8 antibodies be integrated into advanced proteomics workflows?

Integrating MROH8 antibodies into proteomics workflows can provide deeper insights into its function and interactions:

  • Antibody-based proteomics approaches:

    a) Immunoprecipitation-Mass Spectrometry (IP-MS):

    • Use validated MROH8 antibodies to pull down protein complexes

    • Identify interaction partners through LC-MS/MS analysis

    • Compare interactome under different cellular conditions

    • This approach could extend current knowledge of MROH8-TBP interaction to identify additional partners

    b) Reverse Phase Protein Arrays (RPPA):

    • Use highly specific MROH8 antibodies for high-throughput protein quantification

    • Analyze expression across large sample sets

    • Correlate with other signaling proteins simultaneously

    c) Proximity-dependent labeling:

    • Combine with BioID or APEX2 approaches to map proximal proteins

    • Does not require stable interactions, capturing transient associations

  • Antibody-based spatial proteomics:

    a) Multiplexed ion beam imaging (MIBI):

    • Use metal-conjugated MROH8 antibodies

    • Achieve subcellular resolution of protein localization

    • Multiplexed with dozens of other proteins simultaneously

    b) Imaging Mass Cytometry (IMC):

    • Similar to MIBI but using different detection technology

    • Provides spatial context for MROH8 expression in tissue samples

  • Antibody-guided protein structure studies:

    a) Hydrogen-deuterium exchange mass spectrometry (HDX-MS):

    • Use antibodies to stabilize specific protein conformations

    • Probe structural dynamics of MROH8 in different functional states

    b) Cross-linking Mass Spectrometry (XL-MS):

    • Identify proximity relationships within MROH8 complexes

    • Map interaction interfaces at amino acid resolution

  • Integration with single-cell approaches:

    a) Single-cell proteomics:

    • Use MROH8 antibodies in mass cytometry (CyTOF) panels

    • Correlate with other protein markers at single-cell resolution

    b) Spatial transcriptomics integration:

    • Combine with in situ transcriptomics to correlate protein and mRNA levels

    • Address questions about post-transcriptional regulation

These advanced proteomics approaches, when integrated with MROH8 antibodies, can provide comprehensive insights into MROH8's functional roles, interaction networks, and regulatory mechanisms at unprecedented resolution.

What approaches are recommended for generating custom or recombinant antibodies against MROH8?

For researchers requiring specialized MROH8 antibodies beyond commercially available options, several approaches are available:

  • Recombinant monoclonal antibody development:

    a) Phage display technology:

    • Allows selection of high-affinity antibodies against specific MROH8 epitopes

    • Completely animal-free process, addressing ethical concerns

    • Enables antibody optimization through affinity maturation

    • Produces antibodies with superior affinity, sensitivity, and specificity

    b) Single B-cell cloning from immunized animals:

    • Isolate antigen-specific B cells using FACS or microfluidics

    • Amplify antibody variable regions using RT-PCR

    • Clone into expression vectors for recombinant production

    • Recent methods allow rapid generation of human monoclonal antibodies from single antigen-specific antibody secreting cells

    c) Workflow for recombinant antibody production:

    StepDescriptionCritical Parameters
    Antigen designSelect unique MROH8 epitopesAvoid hydrophobic regions; check species conservation
    Library screeningUsing phage, yeast, or mammalian displaySelection stringency; multiple rounds of panning
    Antibody engineeringOptimize framework and CDR regionsStability, solubility, and affinity considerations
    Expression systemHEK293, CHO, or other mammalian cellsCulture conditions, transfection efficiency
    PurificationProtein A/G affinity chromatographyBuffer composition, elution conditions
    ValidationTest specificity and application performanceMultiple validation methods
  • Hybridoma technology with modern improvements:

    • Traditional approach still useful for generating monoclonal antibodies

    • Modern improvements include better screening methods and hybridoma stabilization

    • Sequence the resulting antibodies for recombinant production to ensure reproducibility

  • Antibody engineering and modification approaches:

    • Use existing MROH8 antibodies as starting material

    • Create Fab, F(ab')2, or scFv fragments for specific applications

    • Engineer fusion proteins (e.g., antibody-fluorophore genetic fusions)

    • Generate bispecific antibodies targeting MROH8 and interacting partners

  • Novel affinity reagent development:

    • Nanobodies (VHH fragments): Smaller size, better tissue penetration

    • Aptamers: Nucleic acid-based alternatives to antibodies

    • Designed ankyrin repeat proteins (DARPins): Engineered binding proteins

  • Production and purification optimization:

    • Transient transfection of paired heavy and light chain genes into Expi-HEK293F cells

    • Culture for one week at 37°C with shaking at 125 rpm and 8% CO2

    • Purification using protein A/G affinity chromatography

    • Quality control testing including ELISA, SDS-PAGE, and SEC-HPLC

By utilizing these advanced methods, researchers can develop customized MROH8 antibodies with improved properties for specific research applications, potentially overcoming limitations of commercially available antibodies.

How might MROH8 antibodies be used in translational and clinical research applications?

As our understanding of MROH8's biological functions expands, particularly regarding its potential tumor-suppressive role , MROH8 antibodies may find important applications in translational and clinical research:

  • Biomarker development and validation:

    • Use standardized IHC protocols with validated MROH8 antibodies to develop prognostic biomarkers

    • Correlate expression with patient outcomes in various cancer types beyond pancreatic cancer

    • Develop tissue microarray-based approaches for high-throughput assessment

    • Create companion diagnostics for potential MROH8 pathway-targeting therapies

    • Initial research indicates MROH8 expression correlates with improved survival in pancreatic cancer patients

  • Therapeutic response monitoring:

    • Assess changes in MROH8 expression or localization in response to treatments

    • Develop liquid biopsy approaches if secreted forms or extracellular vesicle-associated forms exist

    • Create multiplexed panels including MROH8 and pathway components (TBP, CAPN2)

    • Longitudinal monitoring during treatment

  • Drug development targeting MROH8 pathways:

    • Use antibodies as tools in high-content screening assays

    • Develop cell-based reporter systems to monitor MROH8 expression

    • Create antibody-drug conjugates targeting MROH8-expressing cells if beneficial

    • Investigate approaches to modulate the METTL16-MROH8-TBP-CAPN2 axis

  • Advancing our molecular understanding of MROH8:

    • Characterize MROH8 expression across tissue types and disease states

    • Identify tissue-specific interaction partners and functions

    • Study post-translational modifications and their functional impacts

    • Investigate MROH8's role in normal physiology and developmental processes

  • Technology development for enhanced detection:

    • Create higher-sensitivity detection methods for circulating MROH8

    • Develop multiplexed approaches to simultaneously assess multiple components of MROH8 pathways

    • Integration with digital pathology and AI-based quantification

    • Point-of-care testing if MROH8 proves clinically relevant

As research continues to uncover MROH8's biological significance, antibodies targeting this protein will remain essential tools for translating fundamental discoveries into clinical applications.

What quality control measures should be implemented when working with MROH8 antibodies?

Implementing rigorous quality control measures is essential for obtaining reliable and reproducible results with MROH8 antibodies:

  • Initial antibody validation:

    Validation ParameterMethodAcceptance Criteria
    SpecificityWestern blot, IP-MSSingle band/peak at expected MW; identified peptides match MROH8
    SensitivityDilution seriesDetection of endogenous protein at recommended dilution
    ReproducibilityMultiple lot testingConsistent performance across antibody lots
    Cross-reactivityTesting against related proteinsNo significant binding to non-target proteins
    Knockout validationKO cell lines or tissuesAbsence of signal in KO samples
  • Routine experimental controls:

    a) Positive controls:

    • Cell lines with confirmed MROH8 expression

    • Recombinant MROH8 protein

    • Tissues known to express MROH8

    b) Negative controls:

    • Primary antibody omission

    • Isotype control antibodies

    • MROH8 knockout or knockdown samples

    • Blocking peptide competition

    c) Technical controls:

    • Loading controls for Western blot

    • Internal reference proteins for IHC/IF

    • Standardized positive samples across experiments

  • Documentation and standardization:

    • Maintain detailed records of:

      • Antibody source, catalog number, lot number

      • Storage conditions and freeze-thaw cycles

      • Dilutions and incubation conditions

      • Sample preparation methods

      • Detection systems and imaging parameters

    • Create standard operating procedures (SOPs) for:

      • Antibody handling and storage

      • Sample preparation for each application

      • Staining/detection protocols

      • Image acquisition settings

      • Data analysis methodology

  • Performance monitoring over time:

    • Regular testing against reference samples

    • Monitoring signal-to-noise ratios

    • Tracking consistency of positive controls

    • Maintaining control charts for quantitative applications

    • Verification upon receipt of new antibody lots

  • Application-specific quality measures:

    a) Western blot:

    • Confirm molecular weight

    • Assess background levels

    • Evaluate signal linearity with loading

    b) IHC/IF:

    • Assess specificity of staining pattern

    • Check background in negative control tissues

    • Confirm reproducibility of subcellular localization

    c) IP/Co-IP:

    • Verify enrichment of target protein

    • Assess non-specific binding

    • Confirm reproducibility of interaction partners

By implementing these quality control measures, researchers can ensure high-quality, reliable data when working with MROH8 antibodies, enhancing the reproducibility and impact of their research.

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.