AIFM2 is a flavoprotein oxidoreductase localized to mitochondria and associated with apoptosis and ferroptosis regulation. The FITC-conjugated antibody enables fluorescent detection of AIFM2 in immunofluorescence (IF) assays, leveraging the fluorophore’s green emission spectrum.
Target Specificity: Recognizes the AIFM2 protein (41 kDa) in human, mouse, and rat samples .
Conjugation: FITC (Fluorescein Isothiocyanate) is covalently linked to the antibody via crosslinking protocols, ensuring stability and fluorescence retention .
Applications: Primarily used in IF, but adaptable to Western blot (WB) and immunohistochemistry (IHC) when combined with secondary detection systems .
Sample Preparation:
Fix cells with 4% paraformaldehyde (15–30 min).
Permeabilize with 0.1% Triton X-100 (10 min).
Blocking:
Primary Antibody :
Visualization:
AIFM2/FSP1 acts as a CoQ-dependent oxidoreductase that inhibits ferroptosis by neutralizing lipid peroxides . Recent studies highlight its role in cancer progression:
Cancer Metabolism: AIFM2 upregulation in radioresistant tumors shifts ferroptosis dependence from GPX4 to FSP1 .
Therapeutic Targeting: Genetic screens identified AIFM2 mutations that sensitize cells to ferroptosis-inducing agents like RSL3 .
AIFM2 shares structural homology with AIFM1 (a key apoptosis effector), but its role in programmed cell death remains less characterized . Emerging data suggest cross-talk between AIFM2 and mitochondrial pathways in stress responses .
Thermofisher Scientific. (2012). FITC-conjugated Antibodies Manual.
Proteintech. (2025). AIFM2/ FSP1 Antibody (20886-1-AP).
Nature. (2023). Integrated chemical and genetic screens unveil FSP1 mechanisms.
Thermo Fisher Scientific. (n.d.). AMID Polyclonal Antibody (PA5-24562).
Antibodies Online. (2019). AIFM2 Antibody (ABIN7244696).
AIFM2 (also known as FSP1 or AMID) is a 41 kDa NAD(P)H-binding oxidoreductase belonging to the FAD-dependent oxidoreductase family. It has gained significant attention for its dual role in cell death pathways - functioning in apoptosis and as a key suppressor of ferroptosis. AIFM2/FSP1 shares sequence similarities with AIFM1 (formerly known as AIF), a mitochondrion-associated enzyme that relocates to the cell nucleus during apoptosis . Recent studies have demonstrated its importance as a parallel glutathione-independent anti-ferroptotic pathway to GPX4, making it a critical target for cancer research. The protein is primarily localized in the cell membrane, nucleus, mitochondria, and cytoplasm, with expression detected in most normal tissues .
AIFM2/FSP1 is a 373 amino acid protein (in its canonical human form) with a molecular weight of approximately 40.5-41 kDa . It functions as an apoptosis-associated flavoprotein with a 6-hydroxy FAD cofactor . The protein has up to two known isoforms and is expressed in most normal tissues as two transcripts of 18 and 40 kb in length. When designing experiments, researchers should note that AIFM2/FSP1 has multiple subcellular localizations (cell membrane, nucleus, mitochondria, and cytoplasm), which can affect detection strategies and experimental interpretation . Its ability to relocate between cellular compartments during stress conditions is critical to its function.
FITC-conjugated AIFM2 antibodies are particularly well-suited for:
For flow cytometry applications, intracellular staining protocols have been successfully validated with human cell lines including A549 cells . When used for immunofluorescence, positive staining has been confirmed in cell lines such as A549, with recommended dilutions between 1:50-1:500 .
Optimal sample preparation depends on the experimental application and cellular context:
For fixed samples:
Fixation: 4% paraformaldehyde (10-15 minutes at room temperature) preserves both protein localization and fluorescence intensity
Permeabilization: 0.1% Triton X-100 (5-10 minutes) allows antibody access to intracellular epitopes
Blocking: 5% normal serum (from the species of secondary antibody origin) for 30-60 minutes reduces background
For flow cytometry:
Cell harvesting should minimize stress that might alter AIFM2 expression
Fixation with 2% paraformaldehyde followed by permeabilization with saponin-based buffers is effective
Single-cell suspensions should be filtered through a 40-70 μm mesh to remove aggregates
The key cellular systems validated for AIFM2 detection include HeLa cells, HepG2 cells, PC-12 cells, and A549 cells, with demonstrated reactivity in human, mouse, and rat samples .
A robust experimental design requires the following controls:
Additionally, researchers should implement technical controls such as single-color controls for compensation in multicolor flow cytometry experiments. When performing co-localization studies, fluorophore bleed-through controls are essential for accurate interpretation.
Protocol optimization should focus on:
Antibody concentration: Titration experiments starting at the manufacturer's recommended 1:50-1:500 dilution range are essential . The optimal concentration balances specific signal with minimal background.
Incubation conditions: Test both temperature (4°C vs. room temperature) and duration (1-24 hours) to maximize signal-to-noise ratio.
Antigen retrieval: For tissue sections, heat-induced epitope retrieval using TE buffer at pH 9.0 has been validated, though citrate buffer at pH 6.0 can serve as an alternative .
Counterstaining: Choose nuclear counterstains that don't overlap with FITC emission spectrum (DAPI or Hoechst recommended).
Mounting medium: Use anti-fade mounting media containing radical scavengers to preserve FITC fluorescence during imaging and storage.
Buffer composition: PBS with 1-2% BSA at pH 7.4-8.0 (optimal for FITC) minimizes non-specific binding while maintaining FITC fluorescence efficiency.
When encountering signal problems, consider the following troubleshooting approaches:
For weak signals:
Increase antibody concentration (begin with manufacturer's recommended range 1:50-1:500 for IF/ICC)
Optimize antigen retrieval (test both TE buffer pH 9.0 and citrate buffer pH 6.0)
Extend incubation time (overnight at 4°C can improve signal for low-abundance proteins)
Use signal amplification systems like tyramide signal amplification
Check for protein degradation during sample preparation
Ensure proper filter sets for FITC detection (excitation ~495nm, emission ~519nm)
For inconsistent results:
Standardize fixation and permeabilization protocols across experiments
Prepare fresh dilutions of antibody for each experiment
Control incubation times and temperatures precisely
Validate antibody lot-to-lot consistency
Implement positive controls with known AIFM2 expression (HeLa, HepG2 cells)
FITC is moderately susceptible to photobleaching, which can be mitigated by:
Minimizing exposure to excitation light:
Reduce illumination intensity and exposure duration
Use neutral density filters
Focus on a different field than the one to be imaged
Employ electronic shutters to control illumination time
Using anti-fade reagents:
Mount samples in commercial anti-fade media containing anti-photobleaching compounds
Add oxygen scavengers like glucose oxidase/catalase systems
Store slides at 4°C in the dark
Optimizing image acquisition:
Use confocal rather than widefield microscopy when possible
Apply binning to reduce required exposure times
Capture FITC images first in multi-channel experiments
Utilize computational approaches like deconvolution to enhance weak signals
When different methods yield conflicting results:
Validate with orthogonal approaches:
Evaluate technical factors:
Consider biological variables:
AIFM2/FSP1 has been identified as a ferroptosis suppressor, providing an opportunity to study this regulated cell death pathway:
Co-localization studies:
Use FITC-AIFM2 antibodies with markers of lipid peroxidation to investigate spatial relationships
Examine membrane localization patterns in ferroptosis-resistant versus sensitive cells
Monitor AIFM2/FSP1 redistribution during ferroptosis induction
Functional investigations:
Correlate AIFM2/FSP1 expression with sensitivity to ferroptosis inducers
Investigate the relationship between AIFM2/FSP1 levels and lipid ROS accumulation
Study the CoQ oxidoreductase activity of FSP1 in ferroptosis suppression
Therapeutic relevance:
Recent publications have demonstrated AIFM2/FSP1's role as a parallel pathway to GPX4 in inhibiting ferroptosis, and upregulation of CoQ shifts ferroptosis dependence from GPX4 to FSP1 in acquired radioresistance .
When designing multicolor panels including FITC-conjugated AIFM2 antibodies:
Panel design considerations:
FITC has moderate brightness and is excited by the 488nm laser
Compensation is critical between FITC and PE due to spectral overlap
Pair with spectrally distinct fluorophores like APC for co-staining
Sample optimization:
Suggested panel for ferroptosis research:
| Target | Fluorophore | Purpose |
|---|---|---|
| AIFM2/FSP1 | FITC | Ferroptosis suppressor protein |
| GPX4 | APC | Primary ferroptosis defense enzyme |
| C11-BODIPY | PE | Lipid peroxidation indicator |
| Annexin V | Pacific Blue | Phosphatidylserine exposure marker |
| 7-AAD | PerCP | Cell viability |
For intracellular staining protocols, fixation and permeabilization are essential, with validated results observed in cell lines such as A549 .
Quantitative analysis of AIFM2 localization requires:
Imaging considerations:
Use confocal microscopy for higher spatial resolution
Employ z-stacks to capture complete cellular volume
Include appropriate subcellular markers (mitochondria, nucleus, plasma membrane)
Maintain consistent acquisition settings across experimental conditions
Quantification methods:
Pearson's or Mander's correlation coefficients for co-localization analysis
Nuclear/cytoplasmic ratio measurements
Membrane/cytoplasmic distribution quantification
Use intensity line profiles across cells to visualize distribution patterns
Interpretation guidelines:
Membrane localization is critical for FSP1's anti-ferroptotic function
Nuclear translocation may indicate involvement in transcriptional regulation
Mitochondrial association suggests potential roles in mitochondrial-dependent cell death
Changes in localization patterns after treatments may indicate mechanism of action
Different experimental methods require specific quantification approaches:
Flow cytometry analysis:
Report median fluorescence intensity (MFI) rather than mean (less affected by outliers)
Use fold change in MFI relative to controls for comparisons
Apply appropriate statistical tests (paired t-tests for treated vs. untreated same cell line)
Immunofluorescence quantification:
Measure corrected total cell fluorescence (CTCF) = Integrated Density - (Area × Mean background fluorescence)
Quantify subcellular distribution using nuclear/cytoplasmic ratios
Analyze at least 30-50 cells per condition across multiple fields
Western blot analysis:
Statistical considerations:
Perform at least three independent biological replicates
Use non-parametric tests when normal distribution cannot be assumed
Apply appropriate multiple comparison corrections
Interpretation of AIFM2/FSP1 data requires context-specific consideration:
Cancer research context:
Increased expression may indicate ferroptosis resistance mechanisms
Alterations in subcellular localization can suggest adaptation to stress
Correlate with patient outcome data when available
Consider the relationship with GPX4 expression (parallel ferroptosis defense)
Cell death pathway analysis:
Monitor AIFM2/FSP1 in response to apoptotic vs. ferroptotic stimuli
Assess relationship between AIFM2/FSP1 and markers of oxidative stress
Evaluate changes in expression during different cell death stages
Consider interactions with other cell death regulators
Therapeutic response studies:
AIFM2/FSP1 upregulation may indicate development of resistance
Changes in localization pattern can suggest mechanism of drug action
Alterations in post-translational modifications might affect function
Consider AIFM2/FSP1 as a potential biomarker for treatment response
Recent publications have demonstrated that upregulation of CoQ oxidoreductase FSP1 contributes to acquired radioresistance by shifting ferroptosis dependence from GPX4 to FSP1 .
To ensure reproducible results:
Antibody validation practices:
Verify specificity using positive and negative controls
Test multiple antibody dilutions to determine optimal range
Document lot number and validate new lots against previous results
Use genetic approaches (siRNA/CRISPR) to confirm specificity
Experimental standardization:
Maintain consistent fixation and permeabilization protocols
Use the same imaging or cytometry settings across experiments
Include internal standards across experimental batches
Document detailed protocols including timing, buffers, and temperatures
Data management:
Establish clear criteria for cell/field selection prior to analysis
Blind analysis where possible to reduce bias
Use automated analysis pipelines to minimize user variability
Report all technical parameters in publications (antibody dilution, exposure times, etc.)
Quality control:
Regularly test FITC-conjugated antibodies for fluorophore integrity
Monitor for consistent staining patterns in positive control samples
Implement periodic testing against validated standards
Store antibodies according to manufacturer recommendations to maintain performance
Successful applications across western blot, immunoprecipitation, immunohistochemistry, and immunofluorescence have been documented for AIFM2 antibodies, with reproducible results in human, mouse, and rat samples .