MIOS Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Compound Overview: MIOS Antibody, FITC Conjugated

MIOS (Missing Oocyte, Meiosis Regulator Homolog) is a 99 kDa protein critical for amino acid sensing via the mTORC1 pathway, functioning as part of the GATOR2 complex . A FITC-conjugated MIOS antibody combines specificity for MIOS with fluorescein isothiocyanate (FITC), enabling fluorescent detection in applications such as immunofluorescence (IF), flow cytometry, and immunohistochemistry (IHC) .

FITC Conjugation Process

FITC binds covalently to lysine residues on antibodies via primary amines. Optimal conjugation ratios range from 10–400 µg FITC per mg antibody to avoid quenching or solubility issues .

Typical Workflow:

  1. Antibody Preparation: Remove sodium azide (inhibits FITC conjugation) .

  2. FITC Activation: Dissolve FITC in anhydrous DMSO (10 mg/mL) .

  3. Conjugation: Incubate antibody with FITC (40–80 µg/mg antibody recommended) for 1 hour at RT .

  4. Purification: Remove unbound FITC via gel filtration or dialysis .

  5. Quality Control:

    • F/P Ratio: Calculate using absorbance at 280 nm (protein) and 495 nm (FITC) .

    • Brightness Validation: Compare conjugates via cell staining to optimize signal-to-noise ratios .

Immunofluorescence and Flow Cytometry

  • IF/ICC: Dilution range 1:125–1:500 for MIOS detection in U2OS cells .

  • Flow Cytometry: FITC conjugates emit at 519 nm (excitation: 488 nm), compatible with argon lasers .

Functional Studies

MIOS antibodies have been used to study:

  • mTORC1 Regulation: MIOS interacts with DEPDC5/SESTRIN2 in nutrient sensing .

  • Cancer Research: Overexpression in HER2/neu-positive cells correlates with targeted therapy resistance .

Comparative Performance in Multiplex Assays

FluorophoreExcitation (nm)Emission (nm)Photostability
FITC492520Moderate
Alexa Fluor® 488495519High

In Vitro Cytotoxicity Assays

  • HER2/neu-Positive Cells: FITC-conjugated antibodies demonstrated dose-dependent cytotoxicity in SKBR3 and BT-474 cell lines, with IC50 values comparable to T-DM1 (Kadcyla®) .

  • Selectivity: No cytotoxicity observed in HER2/neu-negative MDA-MB-231 cells .

Mucosal Vaccine Development

FITC-labeled antibodies targeting M cells induced robust antigen-specific IgG/IgA responses in mice, enhancing survival against botulinum toxin challenges .

Limitations and Optimization Strategies

  • Photobleaching: FITC signal fades rapidly; use anti-fade mounting media .

  • Cross-Reactivity: Anti-IgG (H+L) secondary antibodies may bind non-target immunoglobulins; validate with Fc-specific controls .

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
MIOS antibody; GATOR complex protein MIOS antibody; Missing oocyte meiosis regulator homolog antibody
Target Names
Uniprot No.

Target Background

Function
This antibody, conjugated with FITC, functions as a component of the GATOR subcomplex GATOR2. It plays a role in the amino acid-sensing branch of the TORC1 signaling pathway. The antibody indirectly activates mTORC1 and the TORC1 signaling pathway by inhibiting the GATOR1 subcomplex. Its activity is negatively regulated by the upstream amino acid sensors SESN2 and CASTOR1.
Database Links

HGNC: 21905

OMIM: 615359

KEGG: hsa:54468

STRING: 9606.ENSP00000339881

UniGene: Hs.520215

Protein Families
WD repeat mio family
Subcellular Location
Lysosome membrane.

Q&A

What is MIOS protein and why is it significant for research applications?

MIOS is a critical component of the GATOR2 complex that regulates the mechanistic target of rapamycin complex 1 (mTORC1) in response to amino acid signals. The GATOR2 complex, which consists of five components (WDR24, MIOS, WDR59, SEH1L, SEC13), is required for amino acids to activate mTORC1 . MIOS has a calculated molecular weight of 99 kDa (875 amino acids) and is encoded by the gene with NCBI Gene ID 54468 . Research interest in MIOS has increased significantly due to its role in autophagy regulation and potential implications in cancer therapy development, particularly for glioblastoma .

Methodologically, when working with MIOS antibodies, researchers should consider:

  • Proper storage conditions (typically -80°C for FITC-conjugated antibodies)

  • Validation in specific applications (IF/ICC, ELISA, WB, IHC)

  • Species reactivity (commonly human, mouse, and rat)

  • Background controls using isotype-matched antibodies

What are the optimal protocols for FITC-conjugated MIOS antibody in flow cytometry?

For optimal flow cytometry using FITC-conjugated antibodies including MIOS detection, researchers should follow these methodological guidelines:

  • Cell preparation: Fix cells using flow cytometry fixation buffer and permeabilize with appropriate permeabilization/wash buffer for intracellular targets .

  • Staining concentration: Typical optimal concentrations range from 0.25 μg of antibody per test, though titration experiments are recommended for each new lot of antibody .

  • Staining conditions: Incubate cell suspensions (approximately 1 × 10^6 cells) in PBS containing 0.5% FCS and 0.1% sodium azide at a concentration of 40 μg/ml of primary antibody for 30 minutes on ice .

  • Washing steps: After incubation, wash cells with buffer and if using indirect detection, incubate with secondary antibody (anti-mouse or anti-rabbit IgG-FITC conjugate) for an additional 30 minutes .

  • Data acquisition parameters: FITC is excited at 488 nm with emission detected at 515-545 nm . Implement proper compensation if using multiple fluorochromes.

  • Controls: Always include unstained cells, isotype controls, and single-color controls for multicolor experiments to enable proper gating and interpretation.

How should researchers validate MIOS antibody specificity?

Antibody validation is crucial for ensuring experimental reproducibility and accuracy. For MIOS antibodies, the following methodological approaches are recommended:

  • Western blot validation: MIOS antibodies have been validated in several cell lines including K-562, HeLa, and PC-3 cells, as well as rat liver tissue, showing the expected molecular weight of 99 kDa .

  • Knockout/knockdown validation: Use MIOS knockout or knockdown models as negative controls. Published literature demonstrates the use of MIOS-deficient (mios-) mutant cells to validate antibody specificity and functional effects .

  • Cross-reactivity testing: Test against multiple species if working with non-human models, as reactivity can vary. Current MIOS antibodies show reactivity with human, rat, and mouse samples .

  • Application-specific validation: Different applications (WB, IHC, IP, ELISA) require different validation approaches. For example, IHC may require antigen retrieval optimization with either TE buffer pH 9.0 or citrate buffer pH 6.0 .

ApplicationRecommended DilutionValidated Systems
Western Blot1:500-1:1000K-562, HeLa, PC-3 cells, rat liver tissue
Immunohistochemistry1:50-1:500Mouse skeletal muscle tissue
Immunoprecipitation0.5-4.0 μg for 1.0-3.0 mg of total protein lysateHeLa cells

How can FITC-conjugated MIOS antibodies be utilized to investigate mTORC1 signaling in cancer research?

MIOS antibodies, particularly when FITC-conjugated for visualization, offer powerful tools for investigating mTORC1 signaling dysregulation in cancer. Advanced methodological approaches include:

  • Co-localization studies: FITC-conjugated MIOS antibodies can be used alongside other fluorescently-labeled markers to analyze spatial relationships between MIOS and other GATOR2 components or mTORC1 pathway proteins using confocal microscopy.

  • Functional screening assays: As demonstrated in recent research, MIOS inhibition correlates with reduced glioblastoma (GBM) cell proliferation. Researchers utilized MIOS-dependent assays to screen potential inhibitory compounds, with readouts including cell proliferation and autophagy induction .

  • Translational research applications: In GBM models, MIOS targeting provides a potential therapeutic approach independent of p53 status. This is particularly significant given that drug-resistant cancers are often associated with loss of p53 activity, and MIOS-targeted compounds like Mi3 showed significant inhibition of GBM cell proliferation (37-57% at 5-10 μM) in a MIOS-dependent manner .

  • Mechanistic studies: MIOS antibodies can help elucidate how the GATOR2 complex regulates SESN binding, leading to inhibition of GATOR1 (a negative regulator of Rag proteins) and subsequently mTORC1. Loss of GATOR2 components results in reduced mTORC1 activity, leading to increased autophagy .

What are the technical considerations for site-specific FITC conjugation to MIOS antibodies?

Traditional antibody conjugation methods using amine/thiol-reactive chemistries often result in heterogeneous antibody display with potentially hindered biological activity. Advanced site-specific conjugation approaches provide significant improvements:

  • Enzymatic modification strategy: A two-step enzymatic process can be employed:

    • First, deglycosylation using PNGase F to cleave glycans from Asn297 in the Fc region

    • Second, introduction of an azide-handle at Gln295 using microbial transglutaminase (MTGase)

  • Quantification of conjugation efficiency: UV-spectroscopy can be used to determine the number of FITC molecules per antibody, with optimal ratios typically approaching 1:1 for MIOS antibodies to maintain function while providing adequate fluorescence .

  • Functionality preservation: Site-specific conjugation helps maintain antigen-binding capacity compared to random conjugation methods, which is particularly important for preserving MIOS epitope recognition. This approach has shown success with various antibodies including human IgG, rat IgG2b, and recombinant humanized monoclonal IgG1 .

How does MIOS interact with autophagy pathways and what methodologies are appropriate for studying this relationship?

MIOS has emerged as a critical regulator of autophagy through its role in the GATOR2 complex. Advanced methodological approaches for studying this relationship include:

  • Autophagy flux assays: FITC-conjugated MIOS antibodies can be used alongside autophagy markers like LC3 to monitor changes in autophagy flux in response to MIOS modulation.

  • MIOS inhibitor screening: Research has demonstrated that compounds targeting MIOS (such as Mi3) can induce autophagy by inhibiting mTORC1 activity. This screening approach identified potential therapeutic compounds:

    CompoundEffect on GL261 Cell ProliferationEffect on U87-MG Cell ProliferationMIOS Dependency
    T2A (control)17-19% reduction (5-10 μM)54-58% reduction (p<0.0001)Yes
    Mi337-57% reduction (5-10 μM)25% reduction (p<0.0001)Yes
    Mi10No significant effectNo significant effectN/A
  • Genetic models: The use of mios- mutant cells provides a valuable system for validating the specificity of MIOS-targeting compounds. For example, Mi3 reduced wild-type cell proliferation in a dose-dependent manner (39% reduction at 2.5 μM increasing to 66% at 10 μM), but had no effect on mios- cells, confirming specificity .

  • Pathway analysis: MIOS functions in relation to SESN (sestrin) binding to the GATOR2 complex, leading to inhibition of GATOR1 and subsequent mTORC1 activity. Investigating this pathway requires specialized assays for measuring mTORC1 activity (e.g., phosphorylation of S6K and 4E-BP1) in conjunction with MIOS visualization or modulation .

What are potential pitfalls in multiparameter flow cytometry using FITC-conjugated MIOS antibodies?

When conducting multiparameter flow cytometry experiments with FITC-conjugated antibodies including MIOS detection, researchers should be aware of several methodological challenges:

  • Spectral overlap considerations: FITC emission (515-545 nm) can overlap with other commonly used fluorophores like PE and GFP. Proper compensation is essential, particularly when using multiple fluorochromes in the same experiment .

  • Fixation and permeabilization effects: Different fixation and permeabilization protocols can affect antibody binding and fluorescence intensity. For intracellular targets like MIOS, optimization of fixation/permeabilization conditions is crucial for maintaining both antigen epitope integrity and cellular morphology .

  • Autofluorescence management: Certain cell types exhibit significant autofluorescence in the FITC channel. Background subtraction and appropriate controls (unstained cells, isotype controls) are essential for accurate data interpretation.

  • Antibody titration: Suboptimal antibody concentrations can lead to either poor signal or high background. Titration experiments should determine the optimal concentration where signal-to-noise ratio is maximized specifically for MIOS detection .

  • Cell preparation artifacts: Improper cell preparation can lead to cell aggregation or death, which can affect both binding and fluorescence characteristics. Single-cell suspensions with high viability are essential for reliable results.

  • Sequential staining considerations: When combining surface and intracellular markers, the sequence of staining becomes important. Generally, surface staining should be performed before fixation and permeabilization for intracellular targets like MIOS.

How can MIOS antibodies contribute to therapeutic development for glioblastoma?

Recent research has identified MIOS as a potential therapeutic target for glioblastoma (GBM), with several methodological approaches being developed:

  • Drug screening approach: Using molecular docking against the β-propeller of MIOS (the secondary structure likely involved in binding MIOS into the GATOR2 complex), researchers have identified compounds with strong binding potential (indicated by ΔG values) . This rational drug design approach offers a more targeted strategy than traditional high-throughput screening.

  • Functional validation: Promising compounds can be functionally validated in GBM cell lines (GL261 and U87-MG), with cell proliferation inhibition as a readout for efficacy. This approach has successfully identified Mi3 as a potential MIOS inhibitor with significant anti-proliferative effects .

  • Mechanism-specific targeting: MIOS targeting provides a p53-independent anti-cancer mechanism, which is particularly valuable since approximately 50% of cancer types have dysregulated p53. By targeting MIOS downstream of SESN, researchers can inhibit mTORC1 activity independent of p53 status .

  • Combinatorial approaches: MIOS antibodies labeled with FITC can be used to track MIOS expression and localization in response to combination treatments, potentially identifying synergistic therapeutic strategies.

What methodological advances improve sensitivity when using FITC-conjugated antibodies for low-abundance targets?

For detecting low-abundance targets like MIOS in certain cell types, several advanced methodological approaches can enhance sensitivity:

  • Signal amplification systems: Techniques such as tyramide signal amplification (TSA) can be employed with FITC-conjugated antibodies to enhance signal intensity for low-abundance targets.

  • Optimized fixation and permeabilization: For intracellular targets, specialized fixation and permeabilization protocols can significantly improve antibody penetration and binding while preserving antigen epitopes. For example:

    • For flow cytometry: Use of specialized fixation buffers followed by permeabilization with detergents like saponin or Triton X-100

    • For immunofluorescence: Optimization of fixation time, temperature, and buffer composition

  • High-sensitivity detection systems: Modern flow cytometers with improved photomultiplier tubes (PMTs) and specialized filters can enhance FITC detection sensitivity. Similarly, advanced microscopy techniques like confocal microscopy with photon counting or super-resolution microscopy can significantly improve signal detection.

  • Background reduction strategies: For tissue samples particularly, autofluorescence quenching reagents can improve signal-to-noise ratios for FITC-conjugated antibodies.

  • Antibody fragment utilization: In some applications, using F(ab) or F(ab')₂ fragments conjugated with FITC instead of whole antibodies can improve tissue penetration and reduce non-specific binding through Fc receptors.

How can researchers optimize reproducibility when using FITC-conjugated MIOS antibodies across different experimental systems?

Ensuring reproducibility is a major challenge in antibody-based research. For FITC-conjugated MIOS antibodies, consider these methodological approaches:

  • Standardized antibody validation: Implement a comprehensive validation strategy across different lots using:

    • Western blot confirmation of specificity with proper molecular weight (99 kDa for MIOS)

    • Positive and negative control samples (e.g., MIOS knockout/knockdown models)

    • Cross-platform validation (flow cytometry, microscopy, Western blot)

  • Reference standards: Establish internal reference standards for fluorescence intensity calibration, such as calibration beads with known quantities of FITC molecules.

  • Detailed protocol documentation: Document all experimental parameters including:

    • Antibody source, catalog number, lot number, and concentration

    • Buffer compositions and pH

    • Incubation times and temperatures

    • Instrument settings (laser power, PMT voltage, compensation matrix)

    • Data analysis parameters (gating strategy, background subtraction method)

  • Inter-laboratory validation: When possible, validate key findings across different laboratories using the same antibody lots and protocols.

  • Storage and handling optimization: FITC is susceptible to photobleaching and pH-dependent fluorescence changes. Standardize:

    • Protection from light during all stages

    • Maintenance of slightly alkaline conditions (pH 7.3-8.0) for optimal FITC fluorescence

    • Aliquoting of antibodies to avoid freeze-thaw cycles

    • Storage at recommended temperatures (typically -20°C or -80°C)

How should researchers address non-specific binding when using FITC-conjugated MIOS antibodies?

Non-specific binding can significantly impact data quality. Methodological approaches to address this issue include:

  • Optimized blocking strategies:

    • For immunofluorescence: Use of 5-10% serum from the same species as the secondary antibody

    • For flow cytometry: Inclusion of 1-2% BSA or 5-10% serum in staining buffers

    • For tissues with high Fc receptor expression: Addition of Fc receptor blocking reagents

  • Titration experiments: Determine the optimal antibody concentration that maximizes specific signal while minimizing background. Starting dilution ranges for MIOS antibodies typically range from 1:50-1:500 for IHC and 1:200 for flow cytometry .

  • Isotype control optimization: Use properly matched isotype controls (e.g., Rabbit IgG for polyclonal MIOS antibodies, Mouse IgG2a kappa for monoclonal antibodies) at the same concentration as the primary antibody .

  • Secondary antibody cross-adsorption: For indirect detection methods, use highly cross-adsorbed secondary antibodies to minimize species cross-reactivity.

  • Sample-specific considerations: Different sample types require tailored approaches:

    • Cell lines: Proper fixation/permeabilization optimization

    • Tissue sections: Antigen retrieval method optimization (TE buffer pH 9.0 or citrate buffer pH 6.0 for MIOS)

    • Primary cells: Consideration of autofluorescence reduction methods

What quality control measures should be implemented when working with FITC-conjugated MIOS antibodies?

Rigorous quality control is essential for reliable results with FITC-conjugated antibodies. Key methodological considerations include:

  • Antibody performance metrics:

    • Determination of signal-to-noise ratio in relevant experimental systems

    • Assessment of lot-to-lot variability using standard samples

    • Regular validation of specificity using positive and negative controls

  • Fluorophore quality assessment:

    • Monitoring of FITC:protein ratio using spectrophotometric methods

    • Assessment of free FITC contamination

    • Regular evaluation of photobleaching rates under experimental conditions

  • Instrument quality control:

    • For flow cytometry: Regular calibration using standardized beads

    • For microscopy: Laser power monitoring and point spread function verification

    • Standardized PMT/gain settings for longitudinal studies

  • Sample quality monitoring:

    • Cell viability assessment before fixation

    • Evaluation of morphological preservation after fixation/permeabilization

    • Monitoring of background autofluorescence levels

  • Data analysis standardization:

    • Consistent gating strategies for flow cytometry

    • Standard background subtraction methods for imaging

    • Appropriate statistical analyses for experimental design

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.