MACF1 (microtubule-actin crosslinking factor 1) antibody, FITC conjugated, is a fluorescently labeled polyclonal antibody designed for detecting MACF1 in human, mouse, and rat samples. FITC (fluorescein isothiocyanate) conjugation enables visualization of MACF1 localization and dynamics in cells via fluorescence microscopy, flow cytometry, or immunofluorescence assays . MACF1 is a cytoskeletal linker protein critical for mediating interactions between microtubules and actin filaments, influencing cell migration, polarization, and adhesion .
MACF1 FITC-conjugated antibodies have been used to study:
Preosteoblast migration: MACF1 promotes focal adhesion (FA) turnover by regulating EB1 (end-binding protein 1) distribution on microtubules and FA complexes .
Polarization: MACF1 enhances Golgi-nucleus alignment in migrating cells, as shown in MC3T3-E1 preosteoblasts .
FA turnover: Reduced EB1-FA colocalization in MACF1-overexpressing cells correlates with faster FA disassembly .
In glioblastoma studies, MACF1 inhibition combined with radiation therapy reduced tumor cell viability and migration. FITC-labeled antibodies helped visualize MACF1’s membrane-proximal localization in platelets and glioblastoma cells .
Co-immunoprecipitation (Co-IP) workflows using MACF1 antibodies identified interactions with SMAD7, a regulator of osteogenic differentiation . FITC conjugation enables simultaneous detection of MACF1 and co-localized proteins (e.g., β-catenin, APC) .
Western blot confirmation of a single band at ~600 kDa in human and mouse lysates .
Immunofluorescence in MC3T3-E1 cells showed partial alignment with microtubules, validating cytoskeletal targeting .
MACF1-SMAD7 Interaction: Co-IP and proteomics revealed MACF1 facilitates SMAD7 nuclear translocation, influencing osteoblast differentiation .
Cancer Therapeutic Target: MACF1 knockdown sensitizes glioblastoma cells to radiation by downregulating ribosomal protein S6 .
Cytokinesis Regulation: MACF1 deficiency causes cytokinesis defects and S-phase arrest in osteoblasts, increasing binuclear cells .
MACF1 (microtubule-actin crosslinking factor 1) is a large (approximately 600-620 kDa) spectraplakin family protein that functions as one of the few molecules capable of directly binding both microtubule and actin filament networks . It plays essential roles in cytoskeletal organization and dynamics, particularly in processes requiring coordinated cytoskeletal reorganization . MACF1 is critically involved in:
Cell polarization and directional migration
Focal adhesion turnover and stability
Preosteoblast migration and differentiation
Mesenchymal stem cell (MSC) osteogenic differentiation
Regulation of protein translocation between cytoplasm and nucleus
Recent research has revealed MACF1's significance in bone development, with reduced expression observed in osteoporotic bone specimens, suggesting its potential as a therapeutic target for degenerative bone diseases .
FITC conjugation provides direct fluorescent labeling of the MACF1 antibody, offering several methodological advantages:
Eliminates need for secondary antibody incubation, simplifying immunofluorescence protocols
Enables direct visualization in flow cytometry and fluorescence microscopy
Facilitates multi-color immunostaining when combined with antibodies labeled with spectrally distinct fluorophores
Allows real-time observation of MACF1 localization and dynamics
When working with FITC-conjugated MACF1 antibodies, researchers should note:
FITC has excitation/emission peaks at approximately 495/520 nm (green fluorescence)
FITC is susceptible to photobleaching, requiring appropriate antifade measures
Signal intensity may be lower than amplified detection systems using secondary antibodies
pH sensitivity should be considered (optimal fluorescence at pH >7.0)
Based on validated reactivity data, researchers should consider the following positive controls for MACF1 antibody experiments:
MACF1 expression varies across tissues, with particularly high abundance reported in bone, specifically in mesenchymal stem cells . When evaluating new cell types or tissues, researchers should first confirm MACF1 expression via Western blot before proceeding to more complex applications with FITC-conjugated antibodies.
While specific dilution recommendations for FITC-conjugated MACF1 antibodies may vary by manufacturer, the following ranges provide a starting point based on unconjugated MACF1 antibody validation data:
Optimal dilution is sample-dependent and should be determined through titration experiments. As FITC-conjugated antibodies typically produce lower signal intensity than detection systems using secondary antibodies, researchers may need to use higher concentrations than with unconjugated primary antibodies .
For optimal epitope exposure in fixed tissue sections, the following antigen retrieval methods have been validated:
The effectiveness of antigen retrieval methods can vary depending on fixation protocol, tissue type, and specific epitope targeted by the antibody. When working with FITC-conjugated MACF1 antibodies, researchers should verify that the fluorophore remains stable under the heating conditions used for antigen retrieval, as some epitope retrieval methods may affect fluorescence intensity.
Rigorous experimental design requires appropriate controls to ensure antibody specificity:
Positive Controls:
Include known MACF1-expressing cells (U-251, NIH/3T3) or tissues (human lung)
Run parallel samples with unconjugated MACF1 antibody detected with secondary antibody
Negative Controls:
MACF1 knockdown/knockout samples (using siRNA or CRISPR-Cas9)
Isotype control antibody (matched IgG-FITC conjugate)
Analysis of tissues from MACF1 conditional knockout models (e.g., Macf1^fl/fl, Pf4-Cre)
Technical Controls:
Primary antibody omission (to assess autofluorescence)
Peptide competition assay (pre-incubation with immunizing peptide)
Serial dilution series to demonstrate signal specificity
The conditional knockout mouse model described in the literature (Macf1^fl/fl, Pf4-Cre) provides an excellent specificity control for MACF1 antibodies in megakaryocyte and platelet studies .
MACF1 plays a critical role in focal adhesion (FA) turnover during cell migration. FITC-conjugated MACF1 antibodies can be employed in fixed-cell studies to investigate this process:
Recommended Protocol:
Culture preosteoblasts (e.g., MC3T3-E1) on fibronectin-coated coverslips
Induce migration (wound healing assay or chemotactic gradient)
Fix cells at various timepoints (0, 15, 30, 60 minutes)
Perform triple immunofluorescence:
FITC-conjugated MACF1 antibody
FA marker (e.g., paxillin or vinculin with spectrally distinct fluorophore)
F-actin staining (e.g., rhodamine-phalloidin)
Analyze:
MACF1 localization relative to FAs and actin cytoskeleton
Co-localization with end-binding protein 1 (EB1) at microtubule plus-ends
FA size, number, and turnover rate
Research has demonstrated that MACF1 overexpression increases preosteoblast migration by 50.7±7.6%, while MACF1 knockdown decreases migration by 64.3±4% . MACF1 influences this process by affecting EB1 distribution along microtubules and at focal adhesions, promoting FA turnover through interactions with Src and APC .
MACF1 has been identified as a direct interactor with SMAD7, facilitating its nuclear translocation to initiate osteogenic differentiation . To investigate this interaction:
Co-localization Analysis:
Culture MSCs under standard or osteogenic conditions
Fix and permeabilize cells at different differentiation stages
Co-stain with:
FITC-conjugated MACF1 antibody
SMAD7 antibody with contrasting fluorophore
Nuclear counterstain (DAPI)
Perform confocal microscopy with z-stack acquisition
Quantify co-localization coefficients in cytoplasmic vs. nuclear compartments
Functional Association Studies:
Compare wild-type and MACF1-deficient MSCs
Track SMAD7 nuclear translocation during osteogenic differentiation
Correlate translocation with differentiation markers and mineralization
Perform rescue experiments with MACF1 constructs
Research has shown that MACF1 deletion significantly reduces SMAD7 expression, particularly in the nucleus, suggesting that MACF1 is required for proper SMAD7 nuclear localization and function .
Visualizing MACF1 in dynamic structures presents several technical challenges:
Solutions:
Use spinning disk or laser scanning confocal microscopy to improve signal isolation
Apply deconvolution algorithms to enhance signal resolution
Consider signal amplification techniques compatible with FITC (e.g., tyramide signal amplification)
Optimize fixation to preserve cytoskeletal structures (e.g., pre-extraction protocols)
Solutions:
Employ super-resolution microscopy (STED, STORM, or SIM) to resolve closely associated cytoskeletal elements
Design triple-labeling experiments (MACF1-FITC, tubulin, and F-actin)
Use proximity ligation assay (PLA) to visualize MACF1's interaction with specific cytoskeletal components
Consider live-cell imaging approaches for dynamic association studies
Solutions:
Use domain-specific antibodies to map distinct functions
Compare staining patterns with multiple antibodies recognizing different epitopes
Correlate immunofluorescence with biochemical fractionation data
Design experiments to distinguish between different MACF1 conformational states
When imaging focal adhesion dynamics, researchers should note that MACF1 influences EB1 distribution along microtubules and decreases EB1 localization at focal adhesions, promoting their turnover through interactions with Src and APC .
MACF1 exhibits distinct localization patterns that vary by cell type and functional state. When interpreting immunofluorescence data:
Cell-type specific considerations:
In preosteoblasts: MACF1 associates with microtubules and focal adhesions during migration
In mesenchymal stem cells: MACF1 is predominantly cytoplasmic with some nuclear localization
In platelets: MACF1 appears dispensable for cytoskeletal function, showing different roles compared to other cell types
Functional state assessment:
Compare resting vs. activated/stimulated cells
Track changes during differentiation processes
Consider polarization state in migrating cells
Quantitative analysis approaches:
Measure fluorescence intensity ratios between compartments
Calculate co-localization coefficients with relevant markers
Apply digital image analysis to quantify distribution patterns
Research has demonstrated that in mesenchymal stem cells, MACF1 is primarily expressed in the cytoplasm with some nuclear presence, and this distribution pattern is functionally relevant to its role in facilitating SMAD7 nuclear translocation .
MACF1 has dual functions as both a cytoskeletal crosslinker and a facilitator of protein translocation. To distinguish these roles:
For Cytoskeletal Crosslinking Analysis:
Assess MACF1 co-localization with both microtubules and actin filaments
Quantify cytoskeletal network organization in MACF1-manipulated cells
Measure microtubule dynamics (e.g., growth, catastrophe rates) in relation to MACF1 levels
Examine effects on focal adhesion turnover and stability
For Protein Translocation Analysis:
Track nuclear/cytoplasmic ratios of MACF1-interacting proteins (e.g., SMAD7)
Perform nuclear fractionation followed by Western blot analysis
Use photo-activatable constructs to monitor real-time translocation kinetics
Analyze transcriptional outcomes of successful nuclear translocation
Research findings reveal that MACF1 interacts directly with SMAD7 and facilitates its nuclear translocation, which is essential for initiating osteogenic differentiation pathways . This function appears distinct from MACF1's structural role in cytoskeletal organization.
MACF1 involvement in both cellular differentiation and migration presents an apparent paradox, as these processes often have opposing requirements. FITC-conjugated MACF1 antibodies can help resolve these contradictions:
Temporal Analysis:
Track MACF1 localization throughout the differentiation timeline
Determine whether MACF1 serves sequential functions during commitment phases
Interactome Mapping:
Identify stage-specific MACF1 binding partners using co-immunoprecipitation
Compare interactomes during migration versus differentiation phases
Domain-Specific Functions:
Use domain-specific antibodies to determine which regions mediate different functions
Correlate structural information with functional outcomes
Context-Dependent Signaling:
Analyze MACF1's role in specific signaling pathways (e.g., SMAD7 in osteogenesis)
Determine how environmental cues modify MACF1 function
Research has shown dual but complementary roles: MACF1 promotes preosteoblast migration by mediating focal adhesion turnover , while also facilitating mesenchymal stem cell differentiation by enabling SMAD7 nuclear translocation . These findings suggest that MACF1 sequentially supports migration of progenitor cells and then their subsequent differentiation.
MACF1 has recently been implicated in bone homeostasis, with decreased expression observed in osteoporotic bone specimens . FITC-conjugated MACF1 antibodies could advance this research through:
Diagnostic Applications:
Development of flow cytometry panels to assess MACF1 expression in patient-derived MSCs
Correlation of MACF1 levels with clinical measures of bone density
Identification of MACF1 expression patterns specific to osteoporotic phenotypes
Mechanistic Investigations:
Visualization of altered MACF1-SMAD7 interactions in osteoporotic versus healthy MSCs
Assessment of cytoskeletal organization differences in MACF1-deficient osteoblasts
Tracking of osteogenic differentiation defects related to MACF1 insufficiency
Therapeutic Development:
Screening potential compounds that modify MACF1 expression or function
Monitoring MACF1 restoration in response to osteoporosis treatments
Evaluation of gene therapy approaches targeting MACF1 in preclinical models
Studies have established that conditional knockout of mesenchymal MACF1 attenuates bone mass, bone microarchitecture, and bone formation capability significantly , suggesting that MACF1 restoration could represent a novel therapeutic approach for osteoporosis.
Advanced imaging of MACF1 in complex tissues requires innovative approaches:
Tissue Clearing Techniques:
Apply CLARITY, iDISCO, or CUBIC protocols to achieve optical transparency
Perform deep tissue imaging of FITC-conjugated MACF1 antibody penetration
Create 3D reconstructions of MACF1 distribution across bone tissue architecture
Multiplex Imaging Strategies:
Develop cyclic immunofluorescence protocols compatible with FITC-conjugated antibodies
Combine with tissue mass cytometry (IMC) for high-parameter tissue analysis
Implement DNA-barcoded antibody methods for highly multiplexed detection
Correlative Microscopy:
Integrate fluorescence microscopy with electron microscopy for ultrastructural context
Combine intravital microscopy with post-fixation MACF1 immunostaining
Develop cryo-fluorescence approaches to preserve native protein distribution
Computational Analysis:
Apply machine learning algorithms to identify subtle MACF1 distribution patterns
Develop quantitative analysis pipelines for tissue-level MACF1 assessment
Implement digital pathology approaches for high-throughput analysis
These methodological innovations would be particularly valuable for studying MACF1's role in complex tissues like bone, where its expression and function appear critical for normal development and homeostasis .