FTH1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Overview of FTH1 Recombinant Monoclonal Antibody

The FTH1 recombinant monoclonal antibody is a synthetic antibody engineered to detect the heavy subunit of ferritin (FTH1), a critical protein involved in iron homeostasis. It is produced through in vitro cloning and mammalian cell line expression, enabling precise and reproducible detection of FTH1 across diverse experimental platforms.

Production Process

  1. Cloning and Expression:

    • FTH1 antibody genes are isolated from immunoreactive rabbits or synthesized in vitro.

    • These genes are cloned into plasmid vectors and expressed in mammalian cell lines (e.g., HEK293) using exogenous protein expression systems .

  2. Purification:

    • Antibodies are purified via affinity chromatography (e.g., Protein A/G columns) to achieve >95% purity .

Validation Data

ApplicationMethodTested ReactivityDilution Range
Western Blot (WB)SDS-PAGE analysisHuman, Mouse, Rat 1:500–1:5000
Immunohistochemistry (IHC)Tissue stainingHuman placenta, liver 1:20–1:200
Immunofluorescence (IF)Cell imagingHeLa cells 1:50–1:200
Flow Cytometry (FC)Cell surface detectionHuman, Mouse 1:20–1:200

Role in Iron Metabolism

FTH1 antibodies enable the study of ferritin’s ferroxidase activity, which converts Fe²⁺ to Fe³⁺ for safe intracellular storage . Dysregulation of FTH1 is linked to:

  • Neurodegenerative diseases: Alzheimer’s, Parkinson’s, and neurodegeneration with brain iron accumulation (NBIA9) .

  • Cancer: Modulation of angiogenesis via interaction with kininogen .

  • Ferroptosis: Regulation of cellular iron balance to prevent oxidative stress .

Key Publications

StudyApplicationFindings
Pharmacodynamic Insights into Maresin 1 WB, IFFTH1 mediates ROS-driven apoptosis and ferroptosis in cellular models.
Mitochondrial Iron Metabolism and PD-L1 Regulation WBFTH1 influences mitochondrial iron homeostasis and immune evasion in cancer.

Challenges and Future Directions

  • Specificity Concerns: Cross-reactivity with pseudogenes or paralogs (e.g., FTMT) may require validation .

  • Therapeutic Potential: Exploring FTH1 antibodies as biomarkers for iron overload disorders or targets in cancer therapy .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

This recombinant monoclonal antibody, targeting human and mouse FTH1 (Ferritin Heavy Chain), is produced via a multi-step process. FTH1 antibody genes are initially integrated into plasmid vectors. These vectors are then introduced into host cells for protein expression using established exogenous protein expression techniques. Subsequently, the recombinant FTH1 monoclonal antibody is purified using affinity chromatography. Rigorous validation confirms its suitability for ELISA, Western blotting (WB), Immunohistochemistry (IHC), and Flow Cytometry (FC) applications.

FTH1 plays a crucial role in cellular iron homeostasis. As an iron storage protein, it facilitates the sequestration of excess iron and its controlled release for essential cellular processes. Precise regulation of FTH1 and its interactions within ferritin complexes is vital for maintaining iron balance and preventing iron-related cellular damage.

Form
Liquid
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the order fulfillment method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
Apoferritin antibody; Cell proliferation inducing gene 15 protein antibody; Cell proliferation-inducing gene 15 protein antibody; F HC antibody; Ferritin H subunit antibody; Ferritin heavy chain antibody; Ferritin heavy polypeptide 1 antibody; FHC antibody; FRIH antibody; FRIH_HUMAN antibody; FTH 1 antibody; FTH antibody; FTH1 antibody; FTH1 protein antibody; FTHL 6 antibody; FTHL6 antibody; Iron overload autosomal dominant antibody; MGC104426 antibody; N-terminally processed antibody; OK/SW-cl.84 antibody; PIG 15 antibody; PIG15 antibody; Placenta immunoregulatory factor antibody; PLIF antibody; Proliferation inducing gene 15 protein antibody; Proliferation inducing protein 15 antibody
Target Names
Uniprot No.

Target Background

Function

FTH1 (Ferritin Heavy Chain) stores iron in a soluble, non-toxic, and readily bioavailable form, playing a critical role in iron homeostasis. It possesses ferroxidase activity, oxidizing ferrous iron (Fe2+) to its ferric (Fe3+) form for deposition as ferric hydroxides within the ferritin core. Furthermore, FTH1 participates in iron delivery to cells and mediates iron uptake in developing kidney capsule cells.

Gene References Into Functions

The following publications detail the functional roles of FTH1 across various biological contexts:

  1. Human oligodendrocytes undergo apoptosis when exposed to Sema4A, but this can be mitigated by H-ferritin uptake via the Tim-1 receptor, highlighting FTH1's role in iron-dependent cell survival. (PMID: 29457657)
  2. High serum ferritin levels are associated with prolonged red blood cell transfusion dependence post-hematopoietic stem cell transplantation. (PMID: 29046127)
  3. FTH1's role in the FIH (Factor Inhibiting HIF-1) control of HIF-1 (Hypoxia-Inducible Factor 1) activity is reported. (PMID: 29580991)
  4. In K562 cells, ferritin heavy subunit levels are inversely correlated with H19/miR-675 levels, potentially due to increased ROS production upon FHC silencing. (PMID: 29544765)
  5. In MCF-7 and H460 cells, epithelial-to-mesenchymal transition (EMT), increased migration, and survival are linked to FHC's regulation of iron/ROS metabolism and the CXCR4/CXCL12 axis. (PMID: 28774348)
  6. Ferritin heavy chain is implicated in ovarian cancer stem cell expansion and EMT. (PMID: 27566559)
  7. In vivo magnetic resonance imaging of xenografted neuroblastoma tumors uses FTH1 reporter gene expression controlled by a tet-on switch. (PMID: 27732930)
  8. H-ferritin forms highly stable, insoluble complexes with NCOA4 (Nuclear Receptor Coactivator 4), a process modulated by iron. This has implications for understanding ferritinophagy. (PMID: 28754384)
  9. Recombinant human H-chain ferritin self-assembles into nanocages with a narrow size distribution. (PMID: 26853188)
  10. In myelodysplastic syndromes, increased CD163+ macrophage density and HO1/H-ferritin expression correlate with marrow iron and shorter overall survival. (PMID: 26758041)
  11. Low FTH1-positive tumor cells and microglia/macrophages are associated with poor survival in anaplastic astrocytomas, while high FTL-positive microglia/macrophages have a negative prognostic value. (PMID: 28837569)
  12. No significant relationship exists between serum ferritin concentrations and depressive symptoms in Chinese adults. (PMID: 27611581)
  13. Iron oxidation in homopolymeric H-subunit ferritin (HuHF) proceeds with a 2:1 Fe(II):O2 stoichiometry at low iron levels, generating H2O2. (PMID: 28636371)
  14. Elevated serum ferritin levels are associated with hematologic malignancies. (PMID: 25124580)
  15. Ferritin light and heavy chains are required for neural differentiation of bone marrow-derived mesenchymal stem cells under ELF-EMF exposure. (PMID: 26602884)
  16. Human FTH1 displays a general pro-survival function. (PMID: 26886577)
  17. Increased H-ferritin tissue expression and CD68+/H-ferritin+ cells in lymph nodes correlate with adult-onset Still's disease severity. (PMID: 26540556)
  18. Human H-ferritin exhibits different levels of activity compared to its R. catesbeiana counterpart. (PMID: 26327381)
  19. Mammalian cytosolic ferritins are primarily encoded by two genes, FTH and FTL, with similar structures. (PMID: 26518749)
  20. High cytoplasmic FTH1 is associated with favorable prognosis, while nuclear FTH1 staining is associated with adverse prognosis in triple-negative breast cancer. (PMID: 24742827)
  21. Human mesenchymal stem cells expressing an MRI reporter, human ferritin heavy chain (FTH), exhibit multilineage differentiation ability. (PMID: 25448225)
  22. Iron-independent effects of ferritin in myeloma warrant further investigation. (PMID: 24512320)
  23. Significant differences exist in serum iron and hepcidin levels, but not ferritin, between -582A>G hepcidin polymorphism genotypes. (PMID: 25134646)
  24. High ferritin expression is associated with a malignant phenotype in breast cancer. (PMID: 25265351)
  25. High serum ferritin levels are associated with pancreatic cancer. (PMID: 24446242)
  26. FTH facilitates metabolic adaptation to tissue iron overload. (Review; PMID: 24124891)
  27. Tumor necrosis factor-alpha attenuates starvation-induced apoptosis via ferritin heavy chain upregulation in hepatocellular carcinoma cells. (PMID: 24066693)
  28. Elevated serum ferritin is associated with incident type 2 diabetes in healthy Korean men. (PMID: 24098686)
  29. Children with autism spectrum disorders exhibit significantly lower ferritin levels and higher sleep fragmentation/periodic limb movements. (PMID: 24053984)
  30. Higher serum TNF-alpha, IL-6, and ferritin are associated with intense hepatic steatosis. (PMID: 23892696)
  31. Traumatic brain injury upregulates ferritin-heavy-chain in the pericontusional cortex. (PMID: 23079850)
  32. Increased serum ferritin levels negatively correlate with cardiac function post-myocardial infarction treated with percutaneous coronary intervention. (PMID: 23079089)
  33. In leukemic bone marrow, ferritin H is selectively expressed in erythroid blasts (M6). (PMID: 23611361)
  34. Insights into the metal binding sites of human H-chain ferritin and ferrous ion transport into the nanocage are provided. (PMID: 23344859)
  35. Serum ferritin levels are lower in infectious diseases than in hematologic diseases. (PMID: 23324584)
  36. Ferritin levels and hyperuricemia are independently associated with ALT elevation in obese adolescents. (PMID: 23119080)
  37. Increased ferritin and decreased total antioxidant status indicate increased inflammation in chronic hepatitis C. (PMID: 22760008)
  38. H-ferritin overexpression promotes radiation-induced leukemia/lymphoma. (PMID: 22843505)
  39. The hepcidin:ferritin ratio decreases with increasing fibrosis in chronic liver disease. (PMID: 22676252)
  40. Human FTH1 expression in Hansenula polymorpha, including a FTH1-PTH fusion protein, is studied. (PMID: 22212821)
  41. Ferritin heavy chain expression inhibits apoB-100 secretion from hepatoma cells during HCV infection. (PMID: 22443280)
  42. Survivin inhibition by a peptide derived from the human ferritin heavy chain impedes tumor cell growth. (PMID: 22426960)
  43. Elevated serum ferritin in infants is associated with congenital hypothyroidism. (PMID: 22768652)
  44. Insights into ferritin biomineralization under fever conditions and in biomimetic nanomaterial synthesis. (PMID: 22020807)
  45. FTH1 inhibits JNK signaling pathway activation and binds to Daxx, inhibiting Daxx-mediated apoptosis. (PMID: 21573799)
  46. H-ferritin silencing in human metastatic melanoma cells reduces growth, invasiveness, and cell adhesion. (PMID: 22043922)
  47. A positive correlation exists between reactive oxygen species and serum ferritin levels in myelodysplastic syndrome patients. (PMID: 22117997)
  48. The association of secreted IL-10, FHC, and iron homeostasis in Chlamydia trachomatis-infected HeLa-229 cells is reported. (PMID: 21413929)
  49. FTH1 is a functional target gene of the BACH1 transcription factor (ChIP-seq and knockdown analysis in HEK 293 cells). (PMID: 21555518)
  50. H-ferritin silencing sensitizes tumors to chemotherapy. (PMID: 21385903)
Database Links

HGNC: 3976

OMIM: 134770

KEGG: hsa:2495

STRING: 9606.ENSP00000273550

UniGene: Hs.524910

Involvement In Disease
Hemochromatosis 5 (HFE5)
Protein Families
Ferritin family
Tissue Specificity
Expressed in the liver.

Q&A

What is FTH1 and why is it significant in biomedical research?

FTH1, or Ferritin Heavy Chain, is a critical protein involved in cellular iron homeostasis. It functions primarily as an iron-storage protein, facilitating the storage of excess iron and its controlled release when required for cellular processes. Proper regulation of FTH1 and ferritin complexes is essential for maintaining iron balance and preventing iron-related toxicity within cells .

FTH1 plays a crucial role in several biological processes including energy production, DNA synthesis, and cellular respiration. Its significance extends to various pathological conditions, as dysregulation of FTH1 can lead to iron-related disorders and diseases . The strategic importance of FTH1 in these fundamental cellular processes makes it a valuable target for biomedical research seeking to understand iron metabolism and develop interventions for related disorders.

How are FTH1 recombinant monoclonal antibodies produced?

The production of FTH1 recombinant monoclonal antibodies involves a series of sophisticated molecular biology and protein engineering steps:

  • Gene Extraction and Amplification: The process begins with extraction of FTH1 antibody genes from B cells isolated from immunoreactive organisms (typically rabbits). These genes are then amplified for further processing .

  • Vector Construction and Cloning: The antibody genes are incorporated into plasmid vectors, creating modified constructs that can be introduced into expression systems .

  • Host Cell Expression: These modified plasmid vectors are introduced into suitable host cells (commonly mammalian cell lines such as HEK293) for expression using exogenous protein expression technology. This stage enables the production of functional antibodies in substantial quantities .

  • Purification: Following expression, the FTH1 recombinant monoclonal antibody undergoes purification via affinity chromatography, typically involving Protein A affinity columns for antibodies with Fc regions .

  • Validation: The purified antibodies are then validated for specific applications including ELISA, Western Blotting, Immunohistochemistry, and Flow Cytometry to ensure they correctly bind to the target FTH1 protein .

This recombinant production approach offers advantages over traditional hybridoma technology, including consistent lot-to-lot characteristics, continuous supply capability, and animal-free manufacturing processes for certain antibody variants .

What applications are FTH1 recombinant monoclonal antibodies validated for?

FTH1 recombinant monoclonal antibodies have been validated for multiple laboratory applications, with specific recommended dilutions for optimal performance:

ApplicationRecommended Dilution RangePurpose in Research
Western Blotting (WB)1:500-1:5000Protein detection and quantification in cell/tissue lysates
Immunohistochemistry (IHC)1:20-1:800Visualization of FTH1 localization in tissue sections
Flow Cytometry (FC)1:20-1:200Analysis of FTH1 in individual cells
Immunofluorescence (IF)1:50-1:200Subcellular localization studies
Immunoprecipitation (IP)1:50Isolation of FTH1 and associated proteins
Simple Western™1:10-1:50Automated capillary-based western blotting

These antibodies demonstrate cross-reactivity with FTH1 from multiple species, including human and mouse, making them versatile tools for comparative studies across model organisms . The validation process typically includes positive and negative controls to ensure specificity and high affinity binding to the target protein .

What is the molecular weight and basic structure of FTH1 protein?

FTH1 (Ferritin Heavy Chain) has a molecular weight of approximately 21 kDa as detected in western blotting applications . This corresponds to the expected calculated molecular weight of 21,226 Da based on amino acid sequence analysis .

Structurally, FTH1 functions as part of the ferritin complex, which typically consists of 24 subunits arranged in a spherical shell. These subunits are a mixture of heavy (FTH1) and light (FTL) chains, with the ratio varying depending on the tissue type. The FTH1 subunit contains the ferroxidase center responsible for the oxidation of Fe²⁺ to Fe³⁺, which is essential for iron incorporation into the ferritin complex.

The protein's structural characteristics enable it to fulfill its primary function - storing up to 4,500 iron atoms in a soluble, non-toxic form within its hollow interior. This architecture is crucial for cellular iron homeostasis, allowing for the safe sequestration of potentially harmful free iron and its controlled release when needed for cellular processes.

How can researchers optimize FTH1 antibody performance in challenging experimental conditions?

Optimizing FTH1 antibody performance in challenging experimental conditions requires careful consideration of multiple factors:

For Western Blotting applications with limited protein:

  • Employ signal enhancement systems such as biotin-streptavidin amplification when working with low abundance samples.

  • Optimize primary antibody incubation by extending to overnight at 4°C with dilutions in the mid-range (1:1000) rather than at the extremes of the recommended range .

  • Utilize PVDF membranes rather than nitrocellulose for enhanced protein binding capacity and signal retention.

  • Incorporate low-fluorescence blocking agents to reduce background when using fluorescent secondary antibodies.

For Immunohistochemistry of difficult tissues:

  • Test multiple antigen retrieval methods, comparing heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) versus EDTA buffer (pH 9.0).

  • For formalin-fixed tissues, extend antigen retrieval times and optimize protease digestion parameters if applicable.

  • Consider using tyramide signal amplification systems for tissues with low FTH1 expression.

  • Utilize dilutions in the middle of the recommended range (approximately 1:100) as a starting point .

For Flow Cytometry with challenging cell types:

  • Optimize permeabilization conditions, comparing different detergents (Triton X-100, saponin) at varying concentrations.

  • Extend antibody incubation times to ensure complete penetration in complex samples.

  • Begin with antibody dilutions of approximately 1:100 and adjust based on signal-to-noise ratio .

Researchers should systematically document all optimization parameters and perform appropriate controls, including isotype controls and secondary-only controls, to distinguish specific from non-specific binding patterns.

How do FTH1 recombinant monoclonal antibodies compare to polyclonal alternatives in iron metabolism research?

The choice between recombinant monoclonal and polyclonal antibodies represents an important decision point in iron metabolism research:

Specificity and Reproducibility:
Recombinant monoclonal FTH1 antibodies offer superior lot-to-lot consistency compared to polyclonal alternatives. This is particularly important for longitudinal studies tracking FTH1 expression over time or across different experimental conditions. The controlled production process ensures "superior lot-to-lot consistency, continuous supply, and animal-free manufacturing" capabilities . In contrast, polyclonal antibodies recognize multiple epitopes, which can increase detection sensitivity but may introduce variability between production batches.

Detection of Protein Variants and Complexes:
Monoclonal antibodies recognize specific epitopes, which can be advantageous when investigating particular domains of FTH1. For instance, the Cell Signaling Technology monoclonal antibody is "produced by immunizing animals with a synthetic peptide corresponding to residues near the carboxy terminus of human FTH1 protein" . This specificity allows researchers to distinguish between different functional domains or post-translational modifications of FTH1.

Polyclonal antibodies, which detect multiple epitopes simultaneously, may be more effective at identifying FTH1 in protein complexes or when the protein undergoes conformational changes during iron loading or release processes.

Species Cross-Reactivity:
Many recombinant monoclonal FTH1 antibodies are validated for cross-reactivity with multiple species (human, mouse, rat) , making them suitable for comparative studies across model organisms. When selecting an antibody for research involving multiple species, researchers should verify the cross-reactivity profile matches their experimental design.

Methodological Considerations:
For quantitative applications like western blotting and ELISA, recombinant monoclonal antibodies generally provide more consistent results for absolute quantification of FTH1 levels. For techniques like immunohistochemistry in tissues with low FTH1 expression, polyclonal antibodies may offer advantages in sensitivity, albeit with potential trade-offs in specificity.

What are the critical controls required when using FTH1 recombinant monoclonal antibodies in experimental workflows?

Implementing appropriate controls is essential for ensuring reliable and interpretable results when using FTH1 recombinant monoclonal antibodies:

Essential Negative Controls:

  • Isotype Controls: Include isotype-matched irrelevant antibodies (e.g., rabbit IgG for rabbit-derived anti-FTH1) to identify non-specific binding due to Fc receptor interactions or other antibody class-specific artifacts .

  • Secondary Antibody-Only Controls: Samples processed with secondary antibody but no primary antibody help identify background from the detection system itself.

  • FTH1 Knockout/Knockdown Samples: When available, samples with genetic deletion or suppression of FTH1 provide definitive negative controls for antibody specificity validation.

Critical Positive Controls:

  • Tissues/Cells Known to Express FTH1: Certain cell lines with verified FTH1 expression (such as A549 and HeLa cells, as demonstrated in validation studies) serve as reliable positive controls .

  • Iron-Loaded Samples: Cells or tissues treated with iron compounds to upregulate ferritin expression provide physiologically relevant positive controls.

  • Recombinant FTH1 Protein: Purified recombinant protein at known concentrations establishes detection limits and calibration standards.

Application-Specific Controls:

  • For Western Blotting: Include molecular weight markers to confirm the 21 kDa band corresponds to FTH1 .

  • For Immunohistochemistry/Immunofluorescence: Include competing peptide controls where the antibody is pre-incubated with the immunizing peptide to block specific binding.

  • For Co-localization Studies: Include single-label controls to account for potential spectral overlap in multi-label experiments.

Proper documentation of control experiments according to antibody validation guidelines enhances reproducibility and facilitates troubleshooting when unexpected results occur.

How can researchers distinguish between FTH1 and other ferritin subunits in complex biological samples?

Distinguishing between FTH1 (Ferritin Heavy Chain) and other ferritin subunits, particularly FTL (Ferritin Light Chain), presents a significant challenge in iron metabolism research that requires strategic experimental approaches:

Antibody Selection Strategy:

  • Utilize recombinant monoclonal antibodies specifically targeting unique epitopes of FTH1 not present in other ferritin subunits. For example, antibodies targeting the ferroxidase center, which is present in FTH1 but absent in FTL .

  • Verify antibody specificity through western blotting, confirming detection at the correct molecular weight (21 kDa for FTH1 versus 19 kDa for FTL) .

  • Consider complementary approaches using antibodies raised against synthetic peptides corresponding to unique regions, such as "residues near the carboxy terminus of human FTH1 protein" .

Biochemical Separation Techniques:

  • Implement isoelectric focusing prior to western blotting to separate FTH1 (pI ≈ 5.5) from FTL (pI ≈ 5.8) based on their slightly different isoelectric points.

  • Utilize 2D gel electrophoresis combining isoelectric focusing with SDS-PAGE to distinguish ferritin subunits based on both charge and molecular weight differences.

  • Apply sucrose gradient ultracentrifugation to separate ferritin complexes with different H/L subunit compositions, which display subtle differences in density.

Molecular Biology Approaches:

  • Design PCR primers specific to unique regions of FTH1 mRNA for RT-qPCR analysis to complement protein detection methods.

  • Implement RNA interference specifically targeting FTH1 to create comparative samples with reduced FTH1 expression for antibody validation.

  • Consider immunoprecipitation with anti-FTH1 antibodies followed by mass spectrometry to confirm specificity.

By combining these approaches, researchers can confidently distinguish between ferritin subunits even in tissues where both are expressed, enabling more precise characterization of iron metabolism dynamics under various physiological and pathological conditions.

What are the optimal sample preparation methods for detecting FTH1 in different experimental systems?

The detection of FTH1 requires careful sample preparation tailored to specific experimental systems:

For Western Blotting:

  • Cell Lysate Preparation: Lyse cells in RIPA buffer supplemented with protease inhibitors and iron chelators (such as deferoxamine) to prevent artifactual iron release during processing.

  • Tissue Extraction: Homogenize tissues in cold buffer containing 1% NP-40, 0.5% sodium deoxycholate, and 0.1% SDS with complete protease inhibitor cocktail.

  • Protein Denaturation: Heat samples at 70°C rather than boiling (95-100°C) to prevent ferritin aggregation, which can affect migration patterns.

  • Loading and Transfer: Load 10-30 μg total protein per lane and transfer to PVDF membranes using wet transfer systems for optimal results with the recommended antibody dilutions of 1:1000 for monoclonal antibodies .

For Immunohistochemistry:

  • Fixation: Use 10% neutral buffered formalin for 24-48 hours, as overfixation can mask FTH1 epitopes.

  • Antigen Retrieval: Perform heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes at 95°C.

  • Blocking: Block endogenous peroxidase with 3% hydrogen peroxide followed by protein blocking with 5% normal serum.

  • Antibody Incubation: Apply primary antibody at 1:200-1:800 dilution and incubate overnight at 4°C for optimal staining .

For Flow Cytometry:

  • Cell Fixation: Fix cells with 4% paraformaldehyde for 15 minutes at room temperature.

  • Permeabilization: Permeabilize with 0.1% Triton X-100 for intracellular staining of FTH1.

  • Antibody Staining: Use antibody dilutions of 1:50-1:200 with a minimum 30-minute incubation period .

  • Washing: Perform at least three washing steps between reagent additions to minimize background.

For Immunofluorescence:

  • Cell Culture Preparation: Grow cells on coated coverslips to 70-80% confluence before fixation.

  • Fixation and Permeabilization: Fix with 4% paraformaldehyde for 10 minutes followed by permeabilization with 0.2% Triton X-100.

  • Blocking: Block with 3% BSA in PBS for 1 hour at room temperature.

  • Antibody Application: Apply anti-FTH1 antibody at 1:50-1:200 dilution and counterstain nuclei with DAPI as demonstrated in validation studies using A549 and HeLa cells .

These methodological details ensure optimal detection of FTH1 while minimizing artifacts that could confound data interpretation.

How can researchers quantitatively measure FTH1 levels in various experimental models?

Quantitative measurement of FTH1 requires rigorous methodological approaches across different experimental platforms:

Western Blotting Quantification:

  • Implement linear dynamic range determination for each experimental system using standard curves of recombinant FTH1 protein.

  • Utilize housekeeping proteins like GAPDH or β-actin as loading controls, calculating the ratio of FTH1 to control protein signal.

  • Apply digital image analysis software with background subtraction for densitometric quantification.

  • For precise work, consider running samples at multiple dilutions (neat, 1:2, 1:4) to ensure measurements fall within the linear detection range.

  • Use the recommended antibody dilutions of 1:1000 for standard western blots and 1:10-1:50 for automated capillary-based Simple Western™ systems .

ELISA-Based Quantification:

  • Develop sandwich ELISA systems using capture and detection antibodies targeting different FTH1 epitopes.

  • Generate standard curves using purified recombinant FTH1 protein for absolute quantification.

  • Normalize results to total protein concentration measured by BCA or Bradford assay.

  • Account for matrix effects by preparing standards in the same buffer as experimental samples.

Flow Cytometry Quantification:

  • Use calibration beads with known antibody binding capacity to convert fluorescence intensity to molecules of equivalent soluble fluorochrome (MESF).

  • Apply median fluorescence intensity (MFI) rather than mean values for more robust quantification.

  • Include isotype controls to determine background staining levels.

  • Implement proper compensation controls when performing multiparameter analysis.

  • Use antibody dilutions of 1:50-1:200 as recommended for flow cytometric applications .

Mass Spectrometry-Based Approaches:

  • Develop selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) assays targeting FTH1-specific peptides.

  • Utilize isotopically labeled peptide standards for absolute quantification.

  • Consider immunoprecipitation-mass spectrometry approaches for samples with low FTH1 abundance.

Immunohistochemistry Quantification:

  • Apply digital pathology approaches with trained algorithms to quantify staining intensity and distribution.

  • Use multiple antibody dilutions (1:200, 1:400, 1:800) to establish staining linearity.

  • Include calibration slides with known quantities of target protein for semi-quantitative analysis.

These methodologies enable robust quantitative assessment of FTH1 levels across diverse experimental systems, facilitating comparative studies of iron metabolism regulation.

What strategies can resolve common technical challenges when working with FTH1 antibodies?

Researchers frequently encounter technical challenges when working with FTH1 antibodies that can be addressed through strategic troubleshooting approaches:

Challenge: High Background in Immunohistochemistry and Immunofluorescence

  • Resolution Strategy: Implement a more rigorous blocking protocol using a combination of 5% normal serum and 1% BSA for 1-2 hours at room temperature.

  • Technical Adjustment: Increase washing duration and frequency (5 washes of 5 minutes each) with PBS containing 0.1% Tween-20.

  • Antibody Optimization: Titrate antibody dilutions starting at 1:200 and adjusting based on signal-to-noise ratio .

  • Sample Preparation: Ensure complete deparaffinization of tissue sections and optimize antigen retrieval conditions.

Challenge: Weak or Absent Signal in Western Blotting

  • Resolution Strategy: Verify protein transfer efficiency using reversible total protein staining methods.

  • Technical Adjustment: Optimize lysis conditions to ensure complete solubilization of FTH1, particularly when analyzing iron-loaded samples where ferritin may be less soluble.

  • Antibody Selection: Compare different anti-FTH1 antibodies, as epitope accessibility may vary depending on sample preparation.

  • Signal Enhancement: Implement enhanced chemiluminescence substrates with extended sensitivity or consider fluorescent secondary antibodies for difficult samples.

  • Loading Control: Ensure proper sample loading (10-30 μg total protein) and consider concentrating samples with low FTH1 expression.

Challenge: Cross-Reactivity with Related Proteins

  • Resolution Strategy: Perform validation using knockout/knockdown controls or competing peptide approaches.

  • Technical Adjustment: Increase antibody dilution to reduce non-specific binding while extending incubation time to maintain specific signal.

  • Antibody Selection: Choose antibodies validated against the specific species being studied, confirming cross-reactivity profiles .

  • Data Analysis: Carefully analyze molecular weight patterns, confirming FTH1 detection at the expected 21 kDa position .

Challenge: Variability Between Experiments

  • Resolution Strategy: Standardize all protocols with detailed documentation of critical parameters.

  • Technical Adjustment: Prepare larger volumes of antibody dilutions to use across multiple experiments.

  • Sample Handling: Minimize freeze-thaw cycles of both antibodies and samples.

  • Controls: Include consistent positive controls across experiments for normalization.

  • Antibody Storage: Store antibodies according to manufacturer recommendations, typically at -20°C for long-term storage with 50% glycerol to prevent freeze-thaw damage .

These systematic approaches to common technical challenges enable researchers to generate reliable and reproducible data when working with FTH1 recombinant monoclonal antibodies.

How can researchers validate FTH1 antibody specificity in their experimental models?

Rigorous validation of FTH1 antibody specificity is essential for generating reliable research data. A comprehensive validation strategy should incorporate multiple complementary approaches:

Genetic Validation Approaches:

  • CRISPR/Cas9 Knockout Controls: Generate FTH1 knockout cell lines as definitive negative controls to confirm antibody specificity.

  • siRNA/shRNA Knockdown: Perform transient or stable knockdown of FTH1 expression and confirm reduced signal intensity proportional to knockdown efficiency.

  • Overexpression Controls: Transfect cells with FTH1 expression constructs and verify increased signal detection corresponding to expression levels.

Biochemical Validation Methods:

  • Peptide Competition Assays: Pre-incubate the antibody with the immunizing peptide or recombinant FTH1 protein to block specific binding sites before application to samples.

  • Western Blot Analysis: Confirm detection of a single band at the expected molecular weight of 21 kDa , with signal intensity corresponding to expected FTH1 expression patterns across different tissues or experimental conditions.

  • Immunoprecipitation-Mass Spectrometry: Perform immunoprecipitation with the FTH1 antibody followed by mass spectrometry to identify pulled-down proteins, confirming specific enrichment of FTH1.

Physiological Validation Approaches:

  • Iron Manipulation: Treat cells with iron chelators or iron supplementation to modulate FTH1 expression and verify corresponding changes in antibody signal.

  • Tissue Panel Analysis: Examine antibody staining patterns across tissues with known differential FTH1 expression profiles.

  • Developmental Regulation: Analyze FTH1 detection during developmental stages when iron metabolism undergoes known regulatory changes.

Cross-Validation With Alternative Detection Methods:

  • Correlation With mRNA Expression: Compare protein detection patterns with FTH1 mRNA levels measured by RT-qPCR.

  • Orthogonal Antibodies: Compare staining patterns using multiple antibodies targeting different FTH1 epitopes, such as those directed at "residues near the carboxy terminus of human FTH1 protein" versus other regions.

  • Alternative Detection Technologies: Validate findings using non-antibody-based approaches such as aptamer-based detection or CRISPR-based tagging of endogenous FTH1.

Implementing this multifaceted validation strategy provides confidence in the specificity of FTH1 antibody detection, particularly in novel or complex experimental systems where FTH1 regulation may be subject to unexpected influences.

How can FTH1 antibodies be utilized to investigate iron metabolism disorders?

FTH1 recombinant monoclonal antibodies serve as powerful tools for investigating iron metabolism disorders through multiple research applications:

Diagnostic Biomarker Development:

  • Apply immunohistochemistry protocols (1:200-1:800 dilution ) to analyze FTH1 expression patterns in tissue biopsies from patients with hereditary hemochromatosis, anemia of chronic disease, or neurodegeneration with brain iron accumulation (NBIA).

  • Develop multiplexed immunofluorescence panels combining FTH1 detection with other iron metabolism proteins (transferrin receptor, ferroportin, hepcidin) to create comprehensive iron regulatory profiles.

  • Correlate FTH1 protein levels with disease progression and treatment response to establish prognostic indicators.

Cellular Iron Homeostasis Mechanisms:

  • Implement western blotting (1:500-1:5000 dilution ) to quantify FTH1 expression in response to therapeutic iron chelators or iron supplementation.

  • Apply immunofluorescence (1:50-1:200 dilution ) to track subcellular localization of FTH1 during iron stress or inflammatory stimulation.

  • Utilize flow cytometry to measure single-cell variation in FTH1 expression within heterogeneous cell populations, potentially identifying differential responses to iron-targeted therapies.

Molecular Pathology Studies:

  • Analyze FTH1 iron-loading capacity in neurodegenerative conditions by combining immunoprecipitation with iron quantification assays.

  • Investigate post-translational modifications of FTH1 in pathological states using antibodies specifically recognizing phosphorylated or ubiquitinated forms.

  • Examine the assembly and stability of ferritin complexes in disease states through native gel electrophoresis followed by immunoblotting.

Therapeutic Development and Monitoring:

  • Screen potential therapeutic compounds for their ability to modulate FTH1 expression or function as a biomarker for drug efficacy.

  • Monitor treatment response in iron overload disorders by quantifying changes in FTH1 levels or distribution.

  • Develop companion diagnostic approaches using FTH1 detection to stratify patients for targeted iron metabolism therapies.

The versatility of these antibodies across multiple applications enables researchers to conduct comprehensive investigations into the complex role of FTH1 in iron metabolism disorders, potentially revealing new therapeutic targets and diagnostic approaches.

What are the latest methodological advances in using FTH1 antibodies for intracellular iron imaging?

Recent advances have transformed the application of FTH1 antibodies for intracellular iron imaging, enabling increasingly sophisticated visualization of iron metabolism dynamics:

Super-Resolution Microscopy Approaches:

  • Implement Structured Illumination Microscopy (SIM) with FTH1 antibodies at optimized dilutions (1:50-1:200) to visualize ferritin nanocages below the diffraction limit of conventional microscopy.

  • Apply Stochastic Optical Reconstruction Microscopy (STORM) using FTH1 antibodies conjugated to photoswitchable fluorophores to achieve nanometer-scale resolution of ferritin distribution.

  • Combine these approaches with iron-specific probes to correlate ferritin localization with active sites of iron storage and utilization within subcellular compartments.

Multiplexed Imaging Technologies:

  • Develop multiplexed immunofluorescence panels combining FTH1 detection with organelle markers to map the spatial relationship between ferritin and cellular compartments during iron loading or mobilization.

  • Implement cyclic immunofluorescence (CycIF) to detect up to 40 proteins in the same sample by iterative antibody staining, imaging, and signal removal, allowing comprehensive mapping of the iron interactome.

  • Utilize mass cytometry (CyTOF) with metal-tagged FTH1 antibodies to quantitatively assess ferritin levels alongside dozens of other proteins at single-cell resolution.

Live Cell Imaging Innovations:

  • Engineer cell-permeable FTH1 antibody fragments or nanobodies for live-cell imaging of ferritin dynamics.

  • Combine FTH1 detection with genetically encoded iron sensors to simultaneously visualize the iron-storage protein and free iron pools.

  • Implement fluorescence recovery after photobleaching (FRAP) or photoactivation approaches with tagged FTH1 antibodies to measure ferritin mobility and turnover rates in living cells.

Correlative Light and Electron Microscopy (CLEM):

  • Apply gold-conjugated FTH1 antibodies for transmission electron microscopy to correlate fluorescence signals with ultrastructural features.

  • Implement energy-dispersive X-ray spectroscopy alongside immunogold labeling to simultaneously detect FTH1 protein and elemental iron within ferritin complexes.

  • Develop protocols for in situ cryo-electron tomography of immunolabeled ferritin to visualize native three-dimensional architecture of iron-storage complexes.

These methodological advances significantly enhance researchers' ability to visualize and quantify the dynamics of FTH1-mediated iron storage in both physiological and pathological contexts, offering unprecedented insights into iron metabolism regulation at the subcellular level.

How can researchers use FTH1 antibodies to study the intersection of iron metabolism and inflammation?

The complex relationship between iron metabolism and inflammation represents a critical area of research where FTH1 antibodies provide valuable investigative tools:

Mechanistic Studies of Inflammatory Regulation:

  • Apply western blotting techniques (1:500-1:5000 dilution ) to quantify FTH1 upregulation in response to inflammatory cytokines (IL-6, TNF-α, IL-1β) across different cell types.

  • Implement chromatin immunoprecipitation (ChIP) assays followed by FTH1 protein detection to investigate transcription factor binding to the FTH1 promoter during inflammatory stimulation.

  • Utilize pulse-chase experiments with metabolic labeling and FTH1 immunoprecipitation to measure protein synthesis and degradation rates during inflammatory responses.

Cellular Iron Sequestration During Infection:

  • Apply immunofluorescence microscopy (1:50-1:200 dilution ) to track FTH1 localization in macrophages during bacterial infection, visualizing iron-withholding as an antimicrobial strategy.

  • Implement flow cytometry to quantify FTH1 expression in immune cell populations isolated from infection models or inflammatory disease tissues.

  • Develop co-culture systems with fluorescently tagged pathogens and FTH1 antibody staining to visualize the competition for iron between host cells and microorganisms.

Inflammatory Anemias and Iron-Restricted Erythropoiesis:

  • Analyze bone marrow samples from models of anemia of inflammation using immunohistochemistry (1:50-1:800 dilution ) to evaluate FTH1 expression in erythroid precursors.

  • Combine FTH1 antibodies with markers of erythroid differentiation in flow cytometry panels to identify stages where abnormal iron sequestration impacts red blood cell development.

  • Utilize western blotting to compare FTH1 levels in circulating reticulocytes between healthy controls and subjects with inflammatory conditions.

Translation to Human Inflammatory Diseases:

  • Develop tissue microarrays from inflammatory disease biopsies (rheumatoid arthritis, inflammatory bowel disease, etc.) for high-throughput FTH1 immunohistochemical analysis.

  • Implement multiplexed immunofluorescence to co-localize FTH1 with inflammatory markers and cell-type specific proteins in diseased tissues.

  • Correlate FTH1 expression patterns with disease activity scores and treatment responses to establish potential prognostic biomarkers.

By systematically applying these approaches, researchers can elucidate how inflammation alters iron homeostasis through FTH1 regulation, potentially identifying new therapeutic targets at the intersection of these fundamental biological processes.

What role can FTH1 antibodies play in studying neurodegenerative diseases involving iron dysregulation?

Neurodegenerative disorders frequently involve iron dysregulation, making FTH1 antibodies invaluable tools for investigating these pathological processes:

Brain Region-Specific Iron Metabolism Analysis:

  • Apply immunohistochemistry (1:200-1:800 dilution ) to map FTH1 expression across different brain regions in models of Parkinson's disease, Alzheimer's disease, or Friedreich's ataxia.

  • Implement double-labeling techniques combining FTH1 antibodies with neuronal, glial, or microglial markers to identify cell type-specific alterations in iron storage capacity.

  • Utilize laser capture microdissection of immunostained tissues to isolate specific neuronal populations for downstream molecular analysis of iron regulatory networks.

Cellular Mechanisms of Neurodegeneration:

  • Apply western blotting (1:500-1:5000 dilution ) to quantify FTH1 levels in cellular models exposed to neurodegenerative stimuli (amyloid-β, α-synuclein, oxidative stress).

  • Implement immunofluorescence microscopy to visualize FTH1 co-localization with protein aggregates characteristic of specific neurodegenerative diseases.

  • Develop live-cell imaging approaches using labeled FTH1 antibody fragments to track dynamic changes in ferritin distribution during neuronal stress responses.

Oxidative Stress and Ferroptosis Pathways:

  • Analyze FTH1 expression in neurons undergoing ferroptosis, an iron-dependent form of regulated cell death implicated in neurodegeneration.

  • Implement proximity ligation assays to detect interactions between FTH1 and key proteins involved in ferroptotic pathways.

  • Combine FTH1 detection with markers of lipid peroxidation to correlate iron storage capacity with oxidative stress vulnerability.

Translational Applications:

  • Analyze post-mortem brain tissues from neurodegenerative disease patients to identify disease-specific patterns of FTH1 expression and distribution.

  • Develop methods to detect FTH1 in cerebrospinal fluid as a potential biomarker for iron dysregulation in the central nervous system.

  • Utilize FTH1 antibodies to evaluate the effects of iron chelation therapies or other interventions targeting iron metabolism in preclinical models of neurodegeneration.

These research applications enable detailed investigation of iron metabolism disruptions in neurodegenerative processes, potentially leading to new diagnostic approaches and therapeutic strategies targeting FTH1-mediated iron storage and detoxification mechanisms.

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 2024 Thebiotek. All Rights Reserved.