IFITM1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to IFITM1 Antibody

IFITM1 antibodies are immunological reagents designed to detect and study the IFITM1 protein, a 14–17 kDa transmembrane protein encoded by the IFITM1 gene (chromosome 11p15.5). These antibodies enable researchers to investigate IFITM1's roles in viral restriction, immune cell regulation, and cancer biology .

Protein Structure

  • Domains: Two transmembrane regions (M1, M2) with a conserved intracellular loop (CIL) .

  • Isoforms: Differs from IFITM2/3 by lacking N-terminal extensions but having a C-terminal extension .

  • Post-translational modifications: Palmitoylation and phosphorylation regulate its antiviral activity .

Key Functions

FunctionMechanismCitation
Antiviral activityBlocks viral entry by inhibiting membrane fusion (e.g., HIV-1, HCV, SARS-CoV-2)
Immune regulationModulates CD4+ T-cell differentiation and Th1/Th2 balance
Cancer progressionPromotes breast cancer invasion and aromatase inhibitor resistance via JAK/STAT signaling

Common Uses

  • Western blot (WB): Detects IFITM1 at 14–17 kDa in human, mouse, and rat samples .

  • Immunohistochemistry (IHC): Identifies IFITM1 overexpression in ER+ breast tumors .

  • Flow cytometry: Marks latently HIV-infected CD4+ T cells for antibody-dependent cytolysis .

  • Viral inhibition assays: Quantifies IFITM1's blockade of HCV, EBV, or SARS-CoV-2 entry .

Antiviral Mechanisms

  • HCV: IFITM1 disrupts CD81-HCV interactions, reducing viral entry by 77% .

  • HIV-1: Overexpressed in latently infected cells, enabling targeted antibody-dependent killing .

  • EBV: Competes with viral glycoproteins for EphA2 binding, reducing infection by 70% .

Cancer Studies

  • Breast cancer: IFITM1 knockdown in AI-resistant cells reduces tumor growth by 60% in vivo and upregulates p21 .

  • SCLC: IFITM1 overexpression correlates with distant metastasis in xenograft models .

Protocols and Validation

  • Western blot: Use 15% SDS-PAGE with 20 µg lysate; detect using anti-IFITM1 (1:1,000 dilution) .

  • IHC: Optimized for formalin-fixed paraffin-embedded tissues; score intensity on a 0–2 scale .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary based on the purchasing method or location. For specific delivery details, please contact your local distributors.
Synonyms
9-27 antibody; CD 225 antibody; CD 225 antigen antibody; CD225 antibody; CD225 antigen antibody; Dispanin subfamily A member 2a antibody; DSPA2a antibody; IFI 17 antibody; IFI17 antibody; IFITM1 antibody; IFM1_HUMAN antibody; Interferon induced protein 17 antibody; interferon induced transmembrane protein 1 (9-27) antibody; Interferon induced transmembrane protein 1 antibody; Interferon inducible protein 9-27 antibody; Interferon-induced protein 17 antibody; Interferon-induced transmembrane protein 1 antibody; Interferon-inducible protein 9-27 antibody; Leu 13 antibody; Leu 13 antigen antibody; Leu-13 antigen antibody; LEU13 antibody
Target Names
Uniprot No.

Target Background

Function
IFITM1, an interferon-induced antiviral protein, inhibits viral entry into the host cell cytoplasm by allowing endocytosis but preventing subsequent viral fusion and release of viral contents into the cytosol. It exhibits activity against a wide range of viruses, including influenza A virus, SARS coronaviruses (SARS-CoV and SARS-CoV-2), Marburg virus (MARV), Ebola virus (EBOV), Dengue virus (DNV), West Nile virus (WNV), human immunodeficiency virus type 1 (HIV-1), and hepatitis C virus (HCV). IFITM1 can inhibit viral entry mediated by various viral proteins, including influenza virus hemagglutinin protein, MARV and EBOV GP1,2, and SARS-CoV and SARS-CoV-2 S protein. Moreover, IFITM1 is involved in cell adhesion and regulates cell growth and migration. It also inhibits SARS-CoV-2 S protein-mediated syncytia formation. IFITM1 plays a crucial role in the antiproliferative action of IFN-gamma by inhibiting ERK activation or arresting cell growth in the G1 phase in a p53-dependent manner. It acts as a positive regulator of osteoblast differentiation. In hepatocytes, IFITM proteins work in a coordinated manner to restrict HCV infection by targeting endocytosed HCV virions for lysosomal degradation. IFITM2 and IFITM3 display anti-HCV activity that may complement IFITM1's anti-HCV activity by inhibiting the late stages of HCV entry, possibly through coordinated action by trapping the virion in the endosomal pathway and targeting it for lysosome degradation.
Gene References Into Functions
  1. Strong IFITM1 expression in CD4 T cells in HIV controllers is correlated with immune activation. PMID: 27552157
  2. Using a multi-group heat map from GSE9716 analysis of the GEO database, IFITM1 was identified as a relevant radioresistance gene. Inhibition of IFITM1 in combination with radiotherapy could effectively suppress oral neoplasm cells. PMID: 29770536
  3. Inactivated viral particle inoculation increased the expression of IFITM1 protein at both mRNA and protein levels, inducing an antiviral state in HUVECs. PMID: 29100522
  4. Identification of three distinct mutations that converted IFITM1 or IFITM3 from inhibitors to enhancers of MERS-CoV or SARS-CoV spike protein-mediated entry revealed key structural motifs or residues determining the biological activities of IFITM proteins. PMID: 29263263
  5. Epithelial-mesenchymal transition (EMT) signature was dysregulated by both loss and gain of function of IFITM1, which was partially reverted by Caveolin-1 (CAV1). PMID: 27852071
  6. These findings suggest that IFITM1 protein can restrict alphavirus infection by inhibiting viral fusion with cellular membranes. PMID: 27219333
  7. The transcriptional regulation of IFITM1, 2 and 3 expression has been studied. PMID: 28511927
  8. IFITM1 and IFITM3 inhibit Zika virus infection early in the viral life cycle. PMID: 27268505
  9. IFITM1 suppression blocks proliferation and invasion of aromatase inhibitor-resistant breast cancer in vivo by JAK/STAT1-mediated induction of p21. PMID: 28411130
  10. Compared with CD10, IFITM1 shows superior performance in distinguishing endometrial stroma of adenomyosis from mesenchyma surrounding invasive endometrial adenocarcinoma. PMID: 27124937
  11. Overexpression of IFITM1 is associated with Oral Squamous Cell Carcinomas. PMID: 27221933
  12. High expression of IFITM1 is associated with poor prognosis of colorectal cancer. PMID: 26884876
  13. These findings indicate that overexpression of IFITM1 enhances the aggressive phenotype of triple-negative SUM149 IBC cells, and this effect is dependent on STAT2/BRG1 interaction. PMID: 26897526
  14. It has been proposed that IFITM proteins act in a coordinated manner to restrict HCV infection by targeting endocytosed HCV virions for lysosomal degradation. Studies have shown that the actions of IFITM proteins are indeed virus and cell-type specific. PMID: 26354436
  15. The importance of the C-terminal region of IFITM1 in modulating the antiviral function through controlling protein subcellular localization has been demonstrated. PMID: 25738301
  16. It is suggested that IFITM1 promotes the aggressiveness of colorectal cancer cells via caveolin-1 signaling. PMID: 26259513
  17. In virus-producing cells, IFITMs coalesce with forming virions and are incorporated into HIV-1 viral particles. PMID: 25422070
  18. Incorporation of IFITM1, IFITM2 and IFITM3 into HIV-1 virions impairs viral fusion and spread. PMID: 25464829
  19. Host IFITM3, IFITM2 and IFITM1 facilitate morphogenesis of the human cytomegalovirus assembly. PMID: 25552713
  20. Schizophrenia subjects with higher IFITM mRNA levels in cortical blood vessels have greater disturbances in cortical GABA neurons, suggesting that these cell-type disturbances might be influenced by a shared upstream insult involving immune activation. PMID: 24209773
  21. IFITM1 could be a novel metastasis-promoting gene that enhances the metastatic phenotype in ovarian cancer via epigenetic transcriptional regulation. PMID: 24676393
  22. IFITM1 is a potential valuable addition to immunohistochemical panels used in the diagnosis of cellular mesenchymal uterine tumors. PMID: 24072182
  23. Results suggest that the g.-1920G>A polymorphism in interferon inducible transmembrane protein 1 (IFITM1) may be associated with susceptibility to ulcerative colitis (UC). PMID: 24120510
  24. IFITM1 is essential for the formation of functional blood vessels and stabilizes EC-EC interactions during endothelial lumen formation by regulating tight junction assembly. PMID: 24603679
  25. Although their inhibitory activities were modest when compared to that of tetherin, IFITMs, but not tetherin, directly reduced the expression of HIV-1 proteins including Gag, Vif and Nef. PMID: 23376165
  26. Authors have shown that interferon-induced transmembrane protein 1 (IFITM-1), IFITM-2, and IFITM-3 exhibit a broad spectrum of antiviral activity against several members of the Bunyaviridae family. PMID: 23720721
  27. This study defines IFITM1 as an interferon-stimulated gene effector with action against HCV entry. PMID: 22996292
  28. IFITM1 knockdown in human alveolar-derived bone marrow stromal cells was associated with inhibition of Runx2 mRNA and protein expression. PMID: 22634173
  29. Introduction of anti-miR-130a in hepatocytes increased IFITM1 expression. Hepatocytes stably expressing IFITM1 reduced HCV replication. These findings suggest that HCV infection of hepatocytes upregulates miR-130a. PMID: 22787204
  30. The results suggest that the expression of IFITM1 controls the invasiveness and migration of gastric cancer. PMID: 22609115
  31. Hepatitis C virus infection suppresses the upregulation of a subset of effector molecules, including ISG56 and IFITM1. PMID: 21976647
  32. IFITM1 was expressed in the five human glioma cell lines, and its expressions were positively correlated with their tumorigenicity. PMID: 20838853
  33. IFITM proteins differentially restrict the entry of a broad range of enveloped viruses and modulate cellular tropism independently of viral receptor expression. PMID: 21253575
  34. IFITM1, IFITM2, and IFITM3 inhibit HIV-1 replication through interfering with virus entry. PMID: 21177806
  35. Data show that ULBP1, TFR2 and IFITM1 were associated with increased susceptibility to Vgamma9Vdelta2 T-cell cytotoxicity. PMID: 20220060
  36. Activated IFITM1 is associated with Peutz-Jeghers syndrome polyps. PMID: 20428811
  37. Down regulation of IFITM1 is associated with cervical squamous cell carcinoma. PMID: 20099975
  38. The positive expression level of IFITM1 is associated with the progression of the carcinogenetic process in Peutz-Jeghers syndrome. PMID: 19304549
  39. LEU13 has a novel role different from that in the inhibition of cell proliferation, involved in IFNA-induced refractoriness of RSa cells to X rays. PMID: 12926988
  40. IFITM1 expression profiling could be used for molecular classification of CML, which may also predict survival. PMID: 15661263
  41. Overexpression of 9-27 leads to increased migration and invasiveness by suppressing natural killer cells in gastric cancer. PMID: 15808405
  42. IFITM1 has a role in cellular sensitivity to CDDP in esophageal cancer. PMID: 18202764
  43. IFITM1 plays an important role for the invasion at the early stage of HNSCC progression. PMID: 18829488
  44. Results revealed that the interaction between IFITM1 and CAV-1 could enhance the inhibitory effect of CAV-1 on ERK activation. PMID: 19499152
  45. Antiviral effect of IFITM family members on H1N1 influenza, West Nile virus, and Dengue virus has been studied. PMID: 20064371

Show More

Hide All

Database Links

HGNC: 5412

OMIM: 604456

KEGG: hsa:8519

STRING: 9606.ENSP00000330825

UniGene: Hs.458414

Protein Families
CD225/Dispanin family
Subcellular Location
Cell membrane; Single-pass membrane protein. Lysosome membrane.
Tissue Specificity
Bone (at protein level). Levels greatly elevated in colon cancer, cervical cancer, esophageal cancer and ovarian cancer. Expressed in glioma cell lines.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : WB

Sample type: Human PAM cell

Review: Western blot analysis of IFITMs in PAM cell lines stably expressing Flag-tagged IFITM1, IFITM2, or IFITM3 or CMV alone using an IFITM1-specific antibody and IFITM2-specific antibody against IFITM1 and IFITM2, respectively.

Q&A

What is IFITM1 and what biological role does it play?

IFITM1 (Interferon-Induced Transmembrane Protein 1) is a member of the IFITM family of proteins that function as important host effector molecules in the type I interferon response against viruses. It is primarily expressed following interferon stimulation and acts as a potent antiviral effector against multiple viruses, including hepatitis C virus (HCV). IFITM1 contains two hydrophobic membrane-associated domains (M1 and M2) separated by a conserved intracellular loop (CIL), with distinct N- and C-terminal domains that differentiate it from other IFITM family members . The protein is part of a coordinated cellular immune response that restricts viral infection at early stages, particularly during viral entry. The promoter regions of IFITM genes contain Interferon-Stimulated Response Elements (ISRE) and Gamma-Activated Sequences (GAS), allowing rapid upregulation following interferon signaling .

What is the cellular localization pattern of IFITM1 compared to other IFITM proteins?

IFITM1 demonstrates a distinct localization pattern compared to other family members. While IFITM2 and IFITM3 are predominantly localized to intracellular compartments within the cytoplasm, IFITM1 is mainly distributed at the plasma membrane with some intracellular presence . Immunofluorescence studies using Na⁺/K⁺-ATPase (a plasma membrane biomarker) have confirmed that IFITM1 shows strong colocalization with the plasma membrane, while IFITM2 and IFITM3 appear to reside primarily in cytoplasmic compartments . This distinct localization pattern is crucial for understanding the differential antiviral mechanisms of these proteins, as IFITM1's surface expression allows it to interact directly with viral entry receptors such as CD81 .

What is the membrane topology of IFITM1?

IFITM1 adopts a specific topological orientation in the plasma membrane where the N-terminus points into the cytoplasm and the C-terminus extends extracellularly . This topology has been experimentally verified using IFITM1 constructs with dual tags (HA at the N-terminus and Myc at the C-terminus). When plasma membranes are intact, the N-terminal HA tag cannot be detected by antibodies, but becomes accessible when membranes are permeabilized, confirming that the N-terminus faces the cytoplasm . This topological information is critical for designing appropriate antibodies for different experimental applications and understanding how IFITM1 interacts with viral components during infection.

What are the recommended applications for IFITM1 antibodies in viral research?

IFITM1 antibodies can be effectively employed in multiple research applications to investigate viral restriction mechanisms. Western blotting can quantify IFITM1 expression levels following interferon treatment or viral infection, while immunohistochemistry on paraffin-embedded sections allows visualization of tissue-specific expression patterns . For studying subcellular localization, immunofluorescence microscopy with IFITM1 antibodies can reveal the distribution pattern and potential colocalization with viral entry factors like CD81 . When designing experiments, researchers should select antibodies that target specific regions (N-terminal or C-terminal) depending on the protein's orientation and accessibility in their experimental system. For functional studies, combining antibody-based detection methods with virological assays such as HCVpp (HCV pseudoparticle) entry assays can establish correlations between IFITM1 expression patterns and antiviral activity .

How do I validate the specificity of an IFITM1 antibody for my research?

Validating IFITM1 antibody specificity requires a multi-step approach due to the high sequence homology between IFITM family members. Start with positive and negative controls: use cells treated with type I interferons (which upregulate IFITM1) as positive controls and IFITM1 knockdown cells (using specific shRNA) as negative controls . Western blot analysis should show a single band at the expected molecular weight (approximately 17 kDa), with increased intensity in interferon-treated samples and decreased signal in knockdown cells. Cross-reactivity with IFITM2 and IFITM3 should be assessed using overexpression systems of each individual protein. For immunofluorescence applications, verify specificity by examining cellular localization patterns—IFITM1 should predominantly show plasma membrane staining, unlike the intracellular patterns of IFITM2/3 . Additionally, peptide competition assays using the immunizing peptide (derived from the N-terminal region of human IFITM1) can confirm binding specificity .

What are the important considerations when choosing between N-terminal and C-terminal IFITM1 antibodies?

The choice between N-terminal and C-terminal targeting antibodies should be guided by IFITM1's membrane topology and your experimental design. Since IFITM1's N-terminus faces the cytoplasm while the C-terminus extends extracellularly , this orientation affects epitope accessibility in different applications:

  • For immunofluorescence on non-permeabilized cells: C-terminal antibodies are preferred as they can access the extracellular epitope

  • For co-immunoprecipitation of cytoplasmic interaction partners: N-terminal antibodies may be more effective

  • For detecting post-translational modifications: Select antibodies that don't recognize regions containing known modification sites (such as palmitoylation sites in the CIL domain)

  • For detecting protein-protein interactions: Consider which domain interacts with your protein of interest

Additionally, N-terminal antibodies might better distinguish IFITM1 from IFITM2/3, as IFITM1 lacks the 20-21 amino acid N-terminal extension present in the other family members . When studying membrane-associated complexes, consider whether your antibody might disrupt important protein-protein interactions, particularly as IFITM1 is known to interact with viral entry factors like CD81 .

How can I use IFITM1 antibodies to investigate virus-specific restriction mechanisms?

To investigate virus-specific restriction mechanisms, implement a systematic experimental approach utilizing IFITM1 antibodies. Begin with establishing stable cell lines expressing tagged IFITM1 (e.g., FLAG-tagged) to facilitate detection with high specificity . For viral entry studies, compare HCV pseudoparticle (HCVpp) entry rates between control and IFITM1-expressing cells using luciferase reporter assays, while confirming IFITM1 expression levels via western blotting with antibodies . Fluorescence microscopy combining IFITM1 antibodies with markers for viral entry factors (such as CD81 for HCV) can reveal potential interaction sites. Advanced techniques including Fluorescence Resonance Energy Transfer (FRET) and Proximity Ligation Assays (PLA) can definitively demonstrate physical interactions between IFITM1 and viral components or entry factors . For endosomal escape inhibition studies, employ electron microscopy to visualize viral particles trapped in endosomal compartments in IFITM1-expressing cells . When analyzing different viruses, use virus-specific reporter systems alongside consistent IFITM1 detection methods to enable direct comparison of restriction mechanisms across viral families.

What experimental approaches can resolve contradictory data on IFITM1 function in different viral systems?

Resolving contradictory data on IFITM1 function across viral systems requires a comprehensive experimental strategy addressing multiple variables. First, standardize cell models by establishing stable cell lines expressing equivalent levels of IFITM1 across experiments, verified by quantitative western blotting with calibrated antibody concentrations . Consider cell-type specific factors by comparing IFITM1 localization and function in relevant primary cells versus cell lines, using immunofluorescence to document potential differences in distribution patterns . Systematically investigate post-translational modifications by examining IFITM1 palmitoylation status across cell types and viral infections, as S-palmitoylation is essential for antiviral function . Implement single-cell analysis techniques that combine antibody-based IFITM1 detection with viral infection markers to distinguish cell-to-cell variability from population effects. For time-dependent effects, conduct time-course experiments with synchronized infections and fixed timepoint antibody detection to identify whether contradictions result from temporal dynamics rather than absolute differences. Finally, create site-specific mutants of IFITM1 to identify whether functional residues show virus-specific importance, particularly focusing on the conserved cysteine residues in the CIL domain and the membrane-associated domains .

How do post-translational modifications affect IFITM1 detection with antibodies?

Post-translational modifications (PTMs) can significantly impact IFITM1 detection with antibodies through several mechanisms. S-palmitoylation of conserved cysteine residues, particularly in the conserved intracellular loop (CIL), can alter antibody binding if the epitope includes or is adjacent to these modification sites . This palmitoylation is critical for IFITM1's antiviral function and membrane association, and modified IFITM1 may display altered mobility on SDS-PAGE, appearing at a higher apparent molecular weight than predicted. When using antibodies targeting regions containing PTM sites, consider treating samples with depalmitoylating agents (like hydroxylamine) in parallel experiments to determine whether modification status affects detection sensitivity . Similarly, tyrosine phosphorylation, particularly at conserved tyrosine residues, can interfere with antibody recognition if the phosphorylated residue is within the epitope. For comprehensive analysis, researchers should compare results from antibodies targeting different regions of IFITM1 and consider using phospho-specific antibodies when investigating signaling-dependent regulation. When developing quantitative assays, validate whether your antibody's binding affinity is affected by the modification status of IFITM1 under different cellular conditions, particularly following interferon stimulation which can alter both expression levels and modification patterns .

What are the optimal fixation and permeabilization methods for IFITM1 immunofluorescence studies?

The optimal fixation and permeabilization protocol for IFITM1 immunofluorescence depends on which aspects of IFITM1 biology you're investigating. For plasma membrane localization studies, use 4% paraformaldehyde fixation for 10-15 minutes at room temperature to preserve membrane structure, followed by selective permeabilization experiments: perform parallel staining with and without permeabilization to differentiate between membrane-associated and intracellular pools of IFITM1 . For detecting the topological orientation, use dual-tagged IFITM1 constructs (e.g., HA-IFITM1-Myc) and compare antibody accessibility under permeabilized versus non-permeabilized conditions to confirm that the N-terminus faces the cytoplasm while the C-terminus extends extracellularly . When studying colocalization with membrane proteins like Na⁺/K⁺-ATPase, mild detergent permeabilization with 0.1% Triton X-100 for 5-10 minutes preserves membrane structure while allowing antibody access . For endosomal localization studies comparing IFITM1 with IFITM2/3, methanol fixation at -20°C for 10 minutes may better preserve intracellular compartments. Always validate your fixation method by comparing staining patterns with live-cell imaging of fluorescently-tagged IFITM1 to ensure the fixation process doesn't artificially alter localization patterns.

How can I design experiments to distinguish between direct and indirect antiviral effects of IFITM1?

Designing experiments to distinguish direct from indirect antiviral effects of IFITM1 requires controlling for multiple variables and using complementary approaches. Implement time-of-addition experiments where IFITM1 expression is induced at different stages of the viral life cycle using inducible expression systems, then detect both IFITM1 (using validated antibodies) and viral markers to determine stage-specific inhibition . Use pseudotyped virus particles (like HCVpp) that complete only the entry step of the viral life cycle to isolate entry-specific effects, combined with immunofluorescence to correlate IFITM1 localization with restricted viral entry . For direct interaction studies, employ co-immunoprecipitation with IFITM1 antibodies followed by detection of viral proteins, or use advanced techniques like FRET and PLA to demonstrate physical proximity between IFITM1 and viral components . Create chimeric IFITM proteins by swapping domains between IFITM1 and non-restrictive membrane proteins, then use immunofluorescence to correlate localization patterns with antiviral activity to identify essential functional domains. Control for indirect effects by analyzing the impact of IFITM1 expression on general cellular processes like endosomal acidification using pH-sensitive dyes alongside immunofluorescence detection of IFITM1 . Finally, conduct electron microscopy studies to visualize the physical relationship between IFITM1-positive membranes and viral particles during infection, particularly focusing on endosomal escape events .

What controls should be included when using IFITM1 antibodies in virus restriction studies?

A comprehensive control strategy is essential when using IFITM1 antibodies in virus restriction studies. Include these key controls:

Control TypeDescriptionPurpose
Expression ValidationWestern blot comparing IFITM1 levels in control vs. transduced/transfected cellsConfirms expected expression pattern and antibody specificity
Knockdown ControlCells with shRNA targeting IFITM1Validates signal specificity and establishes baseline virus susceptibility
Interferon ResponseIFN-α/λ treated cells at various concentrationsConfirms natural induction of IFITM1 and physiological relevance
Cross-reactivityParallel detection of IFITM2/3Ensures signals aren't contaminated by detection of related proteins
Functional MutantsIFITM1 with mutations in key functional residues (e.g., palmitoylation sites)Correlates specific protein features with antiviral activity
Localization ControlDual staining with appropriate markers (Na⁺/K⁺-ATPase for plasma membrane)Confirms expected subcellular distribution
Virus SpecificityMultiple viral systems (e.g., HCV, ZIKV, DENV)Determines breadth of restriction activity
Topology ValidationIFITM1 with N- and C-terminal tags under permeabilized/non-permeabilized conditionsConfirms proper membrane orientation

When analyzing results, always quantify both IFITM1 expression levels and viral restriction phenotypes to establish dose-response relationships and account for cell-to-cell variability in expression.

How should researchers interpret changes in IFITM1 localization following viral infection?

When interpreting changes in IFITM1 localization following viral infection, researchers should consider several critical factors. First, establish a clear baseline by documenting the steady-state distribution of IFITM1 in uninfected cells using immunofluorescence, noting that IFITM1 normally exhibits both plasma membrane and some intracellular localization . Following infection, quantify changes in distribution patterns using co-localization analysis with markers for different cellular compartments (plasma membrane, early endosomes, late endosomes, and lysosomes). Relocalization to endosomal compartments may indicate recruitment to sites of viral entry and membrane fusion, supporting a direct antiviral mechanism . Compare the timing of relocalization with known kinetics of viral entry to determine whether changes are preventative or responsive. For accurate interpretation, use time-course experiments to distinguish between redistribution of existing IFITM1 versus synthesis and trafficking of new protein following infection. Consider whether localization changes correlate with altered post-translational modifications, particularly S-palmitoylation which affects membrane association and antiviral function . Also evaluate whether relocalization patterns differ between restrictive and permissive cell types, which may explain cell-type specific differences in antiviral activity. To determine functional significance, create IFITM1 mutants with altered trafficking patterns and assess their antiviral activity compared to wild-type protein.

What analytical approaches can distinguish between expression level effects and functional changes in IFITM1 antiviral activity?

Distinguishing between expression level effects and functional changes in IFITM1 antiviral activity requires sophisticated analytical approaches. Implement dose-response analysis by creating stable cell lines expressing IFITM1 at different controlled levels, then plot viral restriction against protein expression quantified by calibrated western blotting to establish the relationship between abundance and function . Use flow cytometry with dual staining for IFITM1 and viral antigens at the single-cell level to correlate expression with protection, allowing identification of potential threshold effects versus proportional protection. For functional analysis independent of expression level, create cell lines expressing equivalent amounts of wild-type versus mutant IFITM1 (verified by quantitative western blotting), then compare their antiviral activities to identify mutations that specifically affect function without altering expression . Employ biochemical fractionation to determine the proportion of IFITM1 in different cellular compartments, as the functional pool may be a subset of the total detected protein. When analyzing post-translational modifications, calculate modification stoichiometry (proportion of IFITM1 molecules modified) rather than just detecting presence/absence. For mathematical modeling of expression versus function, use partial least squares regression analysis to separate the contributions of expression level, localization pattern, and modification status to the observed antiviral phenotype.

How do I troubleshoot inconsistent IFITM1 antibody staining patterns across experiments?

Inconsistent IFITM1 antibody staining patterns can result from multiple technical and biological variables that must be systematically addressed. Create a standardized protocol with consistent fixation parameters (agent, concentration, duration, and temperature), as overfixation can mask epitopes while underfixation may allow protein redistribution . Optimize antibody dilution by performing titration experiments for each new lot of antibody, as even antibodies from the same manufacturer may vary in optimal working concentration between batches . For membrane proteins like IFITM1, permeabilization conditions significantly impact staining patterns; compare different detergents (Triton X-100, saponin, digitonin) at various concentrations to identify optimal conditions that preserve membrane structure while allowing antibody access . Control for cell cycle and density effects by standardizing seeding density and synchronizing cultures, as IFITM1 localization may vary with these parameters. For interferon-induced expression, establish a consistent protocol for timing and concentration of interferon treatment relative to staining . If using multiple fluorophore-conjugated secondary antibodies, verify there's no bleed-through between channels and that secondary antibodies don't exhibit non-specific binding. Finally, consider epitope masking due to protein-protein interactions or post-translational modifications by comparing different antibodies targeting distinct regions of IFITM1 . Document and standardize image acquisition parameters including exposure time, gain, and post-processing steps to ensure quantitative comparisons between experiments.

What methodological advances are needed to better understand IFITM1 membrane topology and dynamics?

Advancing our understanding of IFITM1 membrane topology and dynamics requires innovative methodological approaches. Super-resolution microscopy techniques (STORM, PALM) coupled with specific IFITM1 antibodies could reveal nanoscale organization within the plasma membrane and potential clustering during viral challenge that conventional microscopy cannot detect . Live-cell imaging using split fluorescent protein systems could monitor IFITM1 topology in real-time during viral entry, with complementary fragments placed at N- and C-terminal domains to confirm their relative orientation across the membrane . Hydrogen-deuterium exchange mass spectrometry combined with antibody epitope mapping could provide detailed structural information about which IFITM1 domains are exposed on each side of the membrane. CRISPR-mediated tagging of endogenous IFITM1 with minimal tags would allow monitoring of native protein dynamics without overexpression artifacts. For studying membrane integration kinetics, pulse-chase experiments using temporally controlled labeling systems coupled with domain-specific antibodies could track how newly synthesized IFITM1 acquires its final topology. Advanced biophysical techniques like solid-state NMR spectroscopy of isotopically-labeled IFITM1 in membrane mimetics could provide atomic-level structural information that antibody-based methods cannot resolve. Implementing these methodologies would resolve current contradictions in the literature regarding IFITM1 topology and its relationship to antiviral function.

How can researchers better integrate IFITM1 antibody data with functional genomics approaches?

Integrating IFITM1 antibody data with functional genomics requires systematic coordination between protein-level studies and genomic/transcriptomic analyses. Develop correlated workflows that couple CRISPR screening data with antibody-based validation, where hits from genome-wide screens for viral restriction factors are systematically validated using IFITM1-specific antibodies to confirm expression changes and localization patterns . Implement single-cell approaches that combine transcriptome analysis with immunofluorescence using IFITM1 antibodies to correlate mRNA expression heterogeneity with protein-level variation and functional outcomes. For regulatory element characterization, couple IFITM1 antibody-based chromatin immunoprecipitation (ChIP) of transcription factors with reporter assays to functionally validate predicted regulatory elements in the IFITM1 promoter region, particularly the ISRE and GAS elements . Create integrated datasets by performing quantitative proteomics on IFITM1 immunoprecipitates alongside RNA-seq analysis following viral infection to correlate changes in IFITM1 protein interactions with transcriptional responses. For structure-function analyses, combine deep mutational scanning of IFITM1 with high-throughput antibody-based phenotyping to comprehensively map functional domains and critical residues . When studying species-specific differences, use cross-species validated antibodies to compare IFITM1 expression patterns across evolutionarily diverse cell types alongside comparative genomics analysis to identify conserved functional elements.

How might single-molecule approaches using IFITM1 antibodies transform our understanding of its antiviral mechanisms?

Single-molecule approaches using IFITM1 antibodies offer transformative potential for understanding antiviral mechanisms at unprecedented resolution. Single-molecule tracking using quantum dot-conjugated Fab fragments derived from IFITM1 antibodies could reveal the dynamics of IFITM1 movement within membranes during viral challenge, potentially identifying restriction hotspots where IFITM1 molecules cluster to prevent viral fusion . Super-resolution techniques like PALM/STORM with specifically labeled antibodies could determine the stoichiometry of IFITM1 in functional antiviral complexes and quantify how this changes during infection. Single-molecule FRET experiments using dual-labeled antibodies could detect conformational changes in IFITM1 structure upon viral binding or during interferon stimulation, potentially revealing activation-dependent structural rearrangements . For studying molecular interactions, single-molecule pull-down assays using surface-immobilized IFITM1 antibodies followed by detection of fluorescently-labeled viral components could determine binding affinities and kinetics of these interactions. Correlative light and electron microscopy combining antibody-based fluorescence with nanoscale structural imaging could visualize the precise relationship between individual IFITM1 molecules and viral particles during entry restriction . Advanced techniques like single-molecule force spectroscopy using antibody-functionalized AFM tips could measure the mechanical properties of IFITM1-containing membranes, potentially revealing how these proteins alter membrane rigidity or curvature to prevent viral fusion.

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.