FN1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Production

FN1 recombinant monoclonal antibodies are produced using in vitro expression systems, where antibody DNA sequences are cloned into controlled genetic vectors. This ensures:

  • Batch-to-batch consistency due to defined genetic sequences .

  • High specificity to FN1 epitopes, such as the 8th type III repeat in the cell-binding region .

  • Scalability for large-scale research or therapeutic use .

Key Attributes:

ParameterDetailsSource(s)
TargetFibronectin 1 (FN1), a glycoprotein critical for ECM assembly, wound healing, and metastasis .
Molecular WeightObserved: ~285 kDa (varies due to glycosylation); Theoretical: ~263 kDa .
ReactivityHuman, Mouse, Rat (cross-species validation in WB, IHC, IF) .
ApplicationsWestern Blot (WB), Immunohistochemistry (IHC), Immunofluorescence (IF), Flow Cytometry .
ClonalityMonoclonal (e.g., Clone 9A5, Clone 568) .

Functional Roles:

  • Binds integrins via the RGD sequence, facilitating ECM fibril formation and cellular processes like embryogenesis .

  • Detects FN1 isoforms in cancer research, particularly in lung carcinoma and renal cell carcinoma .

A. Diagnostic and Therapeutic Utility

  • Cancer Research: Detects FN1 overexpression in non-small cell lung carcinoma (NSCLC) and renal cell carcinoma .

  • QC Reagents: Used to validate rapid diagnostic kits (e.g., Brugia Rapid) via gold-conjugated antibodies .

  • Live-Cell Imaging: Engineered single-chain variable fragments (scFv) enable real-time tracking of post-translational modifications .

B. Comparative Advantages Over Traditional Antibodies

FeatureRecombinant FN1 mAbsTraditional mAbs
ConsistencyHigh batch-to-batch uniformity .Variable due to hybridoma drift .
SensitivityEnhanced signal-to-noise ratio in IF and WB .Moderate background staining .
ScalabilityEasily produced in vitro .Limited by hybridoma culture .

Challenges and Considerations

  • Band Discrepancies: Observed MW (~285 kDa) often exceeds theoretical values (~263 kDa) due to post-translational modifications .

  • Epitope Accessibility: Antibody performance varies based on FN1’s conformational states (soluble vs. matrix-bound) .

  • Storage Stability: Requires -20°C storage with glycerol to prevent aggregation .

Emerging Innovations

  • Phospho-Specific Antibodies: Detect phosphorylated FN1 isoforms in kinase signaling studies .

  • Multispecies Reactivity: New clones (e.g., CSB-MA000296) validated for human, mouse, and rat samples .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the shipping method and destination. Please consult your local distributors for specific delivery timeframes.
Synonyms
CIG antibody; Cold insoluble globulin antibody; Cold-insoluble globulin antibody; DKFZp686F10164 antibody; DKFZp686H0342 antibody; DKFZp686I1370 antibody; DKFZp686O13149 antibody; ED B antibody; Fibronectin 1 antibody; FINC antibody; FINC_HUMAN antibody; FN antibody; FN1 antibody; FNZ antibody; GFND antibody; GFND2 antibody; LETS antibody; Migration stimulating factor antibody; MSF antibody; Ugl-Y3 antibody
Target Names
FN1
Uniprot No.

Target Background

Function
Fibronectins bind to cell surfaces and a variety of molecules, including collagen, fibrin, heparin, DNA, and actin. These proteins play crucial roles in various biological processes such as cell adhesion, motility, opsonization, wound healing, and the maintenance of cell shape. They are involved in osteoblast compaction through the fibronectin fibrillogenesis cell-mediated matrix assembly process, essential for osteoblast mineralization. Fibronectins also participate in the regulation of type I collagen deposition by osteoblasts.

Anastellin, a protein that binds fibronectin, induces fibril formation. This fibronectin polymer, known as superfibronectin, exhibits enhanced adhesive properties. Both anastellin and superfibronectin have been shown to inhibit tumor growth, angiogenesis, and metastasis. Anastellin activates p38 MAPK and inhibits lysophospholipid signaling.
Gene References Into Functions
  1. Studies using primary human coronary artery smooth muscle cells have demonstrated that exposure to hypochlorous acid (HOCl)-modified FN results in decreased adherence, increased proliferation, and altered expression of genes involved in extracellular matrix synthesis and remodeling. PMID: 30237127
  2. Depletion of FN1 by siRNA knockdown significantly reduced the invasive capacity of prostate cancer cells in vitro. PMID: 29391407
  3. Human IL-7 binds more strongly to stretched than to relaxed Fibronectin. PMID: 28845674
  4. TGFB1-mediated PI3K/Akt and p38 MAP kinase dependent alternative splicing of fibronectin extra domain A in human podocyte culture has been reported. PMID: 29729706
  5. Research findings suggest that FN1 plays a prominent role in stimulating glioma growth, invasion, and survival through the activation of the PI3K/AKT signaling pathway. PMID: 30048971
  6. The simultaneous delivery of multiple proinflammatory payloads to the cancer site conferred protective immunity against subsequent tumor challenges. A fully human homolog of IL2-F8-TNF(mut), which retained selectivity similar to its murine counterpart when tested on human material, may open new clinical applications for the immunotherapy of cancer. PMID: 28716814
  7. Under the same condition, p53 protein expression, but not mRNA expression, was reversed by MG132. These findings demonstrate that the level of FN expression is associated with the status and expression of p53 in breast cancer cells. PMID: 28765903
  8. This study demonstrates the dual roles of PTHrP on TGF-b1 signaling and FN up-regulation in glomerular mesangial cells. These data provide new insights to guide the development of therapy for diabetic kidney disease. PMID: 28954822
  9. Data suggest that miR-200b regulates EMT of chemo-resistant breast cancer cells by targeting FN1. Therefore, miR-200b-based therapy may be an effective strategy in treating advanced breast cancer patients. PMID: 28972876
  10. Identification of novel integrin-binding domain mutations in FN1 in patients with glomerulopathy with fibronectin deposits. PMID: 27056061
  11. Fibronectin fragments (FNFr) function as matrikines, driving the chemotactic affinity of prostate cancer cells via the alpha5beta1 integrin. PMID: 27715399
  12. Fn with its inactive compact structure requires unfolding to assemble into active fibrils. Shear stress could induce conformational changes of plasma Fn. PMID: 29470988
  13. B. burgdorferi does not primarily target insoluble matrix Fn deposited on endothelial surfaces but, instead, recruits and induces polymerization of soluble plasma Fn (pFn), an abundant protein in blood plasma that is normally soluble and nonadhesive. PMID: 28396443
  14. miR1271 inhibited glioma cell growth by targeting FN1, and a low level of miR1271 in glioma tumor tissues was associated with lower survival rates in patients with glioma. PMID: 28535003
  15. There is a significant association between a positive fetal fibronectin result and underlying inflammatory pathology of the placenta, even more so than the recognized relationship with short cervical length. PMID: 28535404
  16. This article summarizes the many binding partners for fibronectin, including extracellular matrix proteins, growth factors, and synthetic binding partners, with a particular interest in binding partners whose adhesiveness is impacted by the molecular conformation of the fibronectin fibers. (Review) PMID: 27496349
  17. FN1 fibrils regulate TGFB1-induced epithelial-mesenchymal transition. PMID: 28109697
  18. Breast cancer cells alter the dynamics of stromal fibronectin-collagen interactions. PMID: 27503584
  19. This study suggests that high a1-antitrypsin (AAT) expression might be a negative prognostic marker for lung adenocarcinoma. AAT promoted lung adenocarcinoma metastasis, whose functional target may be fibronectin. These findings provide new insights into the mechanisms of lung adenocarcinoma metastasis. PMID: 28440399
  20. Data show that the expression of ED-B fibronectin was much higher in mesenchymal than prostate cancer cells even after the epithelial to mesenchymal transition. Epithelial to mesenchymal transition is a key step for tumor progression contributing to the metastatic spread. Therefore, circulating cancer cells could seed into the metastatic niche taking advantage of the ED-B fibronectin that they secrete. PMID: 27902486
  21. Thrombomodulin (TM) promotes angiogenesis by enhancing cell adhesion, migration, and FAK activation through interaction with fibronectin. PMID: 27602495
  22. Thyroid nodule stiffness is correlated with fibrosis and expression of Gal-3 and FN-1. PMID: 27809694
  23. EGF and TNFalpha cooperatively promoted the motility of HCC cells mainly through NF-kappaB/p65 mediated synergistic induction of FN in vitro. These findings highlight the crosstalk between EGF and TNFalpha in promoting HCC, and provide potential targets for HCC prevention and treatment. PMID: 28844984
  24. Analysis of FN in breast cancer reveals its role and diagnostic potential. PMID: 27250024
  25. RT-PCR together with Sanger sequencing verified the presence of the FN1-ALK fusion transcripts. PMID: 27469327
  26. Fibronectin is readily modified by ONOOH at low (physiologically-relevant) molar ratios of oxidant to protein. PMID: 27396946
  27. The 45 kDa gelatin-binding domain of fibronectin is responsible for the binding to TGM2. PMID: 27394141
  28. A proteomics study showed a strong association of FN1, A2M, C4BPA, and CFB in molecular subtypes of breast cancer. The findings also revealed that altered level expressions of these selected proteins could classify BC subtypes through plasma and tissue-based expression analysis. PMID: 27498393
  29. FN1/CCL2 levels are elevated in the bronchoalveolar lavage fluid from pulmonary sarcoidosis patients. PMID: 27259755
  30. Cancer-associated fibroblasts organize the fibronectin matrix and promote directional prostate cancer cell migration. PMID: 29021221
  31. FN1 mutations that cause defective fibronectin secretion are found in SMD. PMID: 29100092
  32. FN1 overexpression is an important determinant of thyroid cancer aggressiveness. PMID: 27173027
  33. Thyroid hormone T3 induces fibronectin and HIF-1alpha synthesis via the PI3K/AKT signaling pathway. PMID: 28974422
  34. Mutations in FN are associated with glomerulopathy, but when mutant proteins were studied, the single-nucleotide mutations had only minor effects on conformation and matrix assembly. These mutations may destabilize their FNIII domains or generate dimers of dimers by disulfide cross-linking. PMID: 28745050
  35. Fibronectin and Hepatocyte Growth Factor were shown to be produced by lung fibroblasts and, furthermore, to enhance malignant pleural mesothelioma cell migration and invasion. PMID: 28476806
  36. Study identifies four likely Tourette disorder risk genes with multiple de novo damaging variants in unrelated probands: WWC1 (WW and C2 domain containing 1), CELSR3 (Cadherin EGF LAG seven-pass G-type receptor 3), NIPBL (Nipped-B-like), and FN1 (fibronectin 1). PMID: 28472652
  37. Fibrillar fibronectin on this polymer, but not a globular conformation obtained on control polymers, promotes synergistic presentation of integrin-binding sites and bound bone morphogenetic protein 2 (BMP-2), which enhances mesenchymal stem cell osteogenesis in vitro and drives full regeneration of a nonhealing bone defect in vivo at low GF concentrations. PMID: 27574702
  38. Fn plays a critical role in inflammasome-activated cells by amplifying caspase-1 activation and inducing inflammatory cell death. PMID: 27870323
  39. If the expression of Capon is decreased, myeloma cells are adhered to fibronectin or bone marrow stromal cells (bone marrow mesenchymal stem cells). In addition, the sensitivity of the cell line to chemotherapeutic agents was reduced after silencing Capon in the myeloma cell line which was adhered to bone marrow mesenchymal stem cells. PMID: 28671047
  40. Protease sensitivity resulting from mutations in the Fn-binding sequence could lead to degradation of type I collagen and early embryonic lethality. PMID: 27799304
  41. C-terminal truncation of transglutaminase 2 (TG2) reduces binding to the small intestinal extracellular matrix (ECM) despite retained fibronectin (FN)-binding capacity. PMID: 27685605
  42. Analysis of novel functions for two fibronectin isoforms and the mediating receptors in osteoblast differentiation. PMID: 28325836
  43. In vitro binding assays with purified components reveal that Tie-integrin recognition is direct, and further demonstrate that the receptor binding domain of the Tie2 ligand Ang-1, but not the receptor binding domain of Ang-2, can independently associate with a5b1 or aVb3. Cooperative Tie/integrin interactions selectively stimulate ERK/MAPK signaling in the presence of both Ang-1 and fibronectin. PMID: 27695111
  44. Results directly implicate the heparin-binding sequence of the first type III repeat of fibrillar fibronectin (FNIII1) in realignment of stress fibers in HUVECs and, importantly, show that the matricryptic heparin-binding RWRPK sequence located in FNIII1 is required for the response. PMID: 27521419
  45. TGFbeta elevated the expression of CamK IIbeta and CamK IIdelta, while siRNA silencing of those two subtypes significantly reduced TGFbeta-mediated expression of collagen A1 and fibronectin 1. PMID: 28130256
  46. Findings suggest that the up-regulated level of EDA+ FN is associated with liver damage in nonalcoholic fatty liver disease. PMID: 28397039
  47. In vitro binding studies support a previously unreported two-state "catch-clamp" mechanism of Fn binding by CshA, in which the disordered N-terminal domain of CshA acts to "catch" Fn, via formation of a rapidly assembled but also readily dissociable pre-complex, enabling its neighboring ligand binding domain to tightly clamp the two polypeptides together. PMID: 27920201
  48. These data add new evidence that thermodynamic stability correlates primarily with unfolding rate rather than folding rate. The study also has implications for the question of whether opening of FNIII domains contributes to the stretching of fibronectin matrix fibrils. PMID: 27909052
  49. A positive fFN was associated with preterm birth <32 weeks (15.6% versus 4.2%, p = 0.043), <35 weeks (37.5% versus 11.1%, p = 0.002), <37 weeks (65.6% versus 20.8%, p < 0.001), and earlier gestational ages at delivery (35.2 +/- 3.9 versus 37.4 +/- 2.9, p = 0.001). PMID: 26782923
  50. FN1 plays a role in the development of cisplatin resistance in non-small cell lung cancer (NSCLC), possibly by modulation of beta-catenin signaling through interaction with integrin-beta1 in NSCLC. PMID: 27207836

Show More

Hide All

Database Links

HGNC: 3778

OMIM: 135600

KEGG: hsa:2335

UniGene: Hs.203717

Involvement In Disease
Glomerulopathy with fibronectin deposits 2 (GFND2)
Subcellular Location
Secreted, extracellular space, extracellular matrix.
Tissue Specificity
Expressed in the inner limiting membrane and around blood vessels in the retina (at protein level). Plasma FN (soluble dimeric form) is secreted by hepatocytes. Cellular FN (dimeric or cross-linked multimeric forms), made by fibroblasts, epithelial and ot

Q&A

What is FN1 and why is it significant in biomedical research?

Fibronectin (FN1) is a multifunctional glycoprotein present in the extracellular matrix of tissues. It plays critical roles in cell adhesion, tissue development, wound healing, blood clot formation, and cell signaling pathways. As a key structural component, fibronectin maintains tissue integrity and supports various cellular activities . The protein exists in multiple forms - a soluble dimeric form secreted by hepatocytes in plasma, and dimeric or cross-linked multimeric forms produced by fibroblasts, epithelial cells, and other cell types that are deposited as fibrils in the extracellular matrix . Due to its involvement in numerous physiological and pathological processes, FN1 is an important research target in cancer, cardiovascular disease, developmental biology, stem cell research, and signal transduction studies .

What distinguishes recombinant monoclonal antibodies against FN1 from conventional monoclonal antibodies?

Recombinant monoclonal antibodies against FN1 are synthetically generated in vitro, starting with the harvest of FN1 antibody genes from immunoreactive B cells. These genes are amplified, cloned into phage vectors, and introduced into mammalian cell lines for functional antibody production . In contrast, conventional monoclonal antibodies are typically produced through the traditional hybridoma technique, where antibody-producing B cells are fused with myeloma cells to create hybridomas that secrete antibodies continuously .

The recombinant approach offers several advantages:

CharacteristicRecombinant AntibodiesConventional Monoclonal Antibodies
Production consistencyHigh batch-to-batch reproducibilityMay show batch variation
Supply reliabilityContinuous supply once genes are isolatedDependent on hybridoma stability
CustomizationEasier genetic manipulation for enhancing specificityLimited post-production modification
Animal useReduced animal use after initial gene harvestRequires ongoing animal immunization
ManufacturingAnimal-free for most production phasesAnimal-dependent process

Recombinant antibodies demonstrate superior lot-to-lot consistency and provide a more sustainable supply chain for long-term research projects .

How should researchers select the appropriate FN1 antibody for their specific experimental needs?

Selection of the appropriate FN1 antibody depends on multiple experimental factors:

  • Target species reactivity: Verify the antibody's reactivity with your species of interest. Available FN1 antibodies show varying cross-reactivity with human, mouse, and rat samples .

  • Application compatibility: Confirm the antibody has been validated for your intended application:

    • Western blotting (WB): Most FN1 antibodies work at dilutions between 1:500-1:2000

    • Immunohistochemistry (IHC): Typically used at 1:50-1:400 dilutions

    • Immunofluorescence (IF): Often effective at 1:100-1:800 dilutions

    • Flow cytometry (FC): Usually requires 1:50-1:200 dilutions

    • Functional studies: Some antibodies like HFN 7.1 have function-blocking capabilities

  • Epitope specificity: Consider which domain of FN1 you need to target. Some antibodies recognize specific regions, such as HFN 7.1 which targets the region between the PHSRN synergy and RGD sites spanning type III repeats 9 and 10 .

  • Clonality and isotype: Mouse IgG1 is common for many FN1 monoclonal antibodies, but rabbit IgG alternatives are also available .

  • Validation data: Assess the quality of validation data provided by manufacturers, including verified samples (e.g., HeLa cells, rat liver, human appendix) and observed molecular weight (typically 262-285 kDa) .

What is the optimal protocol for Western blot analysis using FN1 antibodies?

A standardized Western blot protocol for FN1 detection should account for the large molecular weight of the protein (262-285 kDa) and potential multiple isoforms:

  • Sample preparation:

    • Use appropriate lysis buffers containing protease inhibitors

    • Load 20-50 μg of total protein per lane

  • SDS-PAGE optimization:

    • Use low percentage (6-8%) gels to resolve high molecular weight FN1

    • Run the gel at lower voltage (80-100V) for better resolution of large proteins

  • Transfer conditions:

    • Employ wet transfer at 4°C overnight at low voltage (30V)

    • Use PVDF membrane with 0.45 μm pore size for larger proteins

  • Antibody incubation:

    • Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature

    • Dilute primary FN1 antibody at 1:500-1:2000 in blocking buffer

    • Incubate overnight at 4°C with gentle agitation

    • Wash 3-5 times with TBST

    • Use appropriate HRP-conjugated secondary antibody at 1:2000-1:5000

    • Incubate for 1 hour at room temperature

    • Wash thoroughly before detection

  • Detection and analysis:

    • Use enhanced chemiluminescence with exposure times optimized for your sample

    • Be prepared to observe a band around 285 kDa, which may differ from the calculated MW (263 kDa) due to post-translational modifications

    • Multiple bands may be detected due to alternative splicing (FN1 has three regions subject to alternative splicing with potential for 20 different transcript variants)

How can researchers optimize immunohistochemistry protocols for FN1 detection in tissue samples?

For optimal FN1 detection in tissue sections:

  • Tissue preparation:

    • Fix tissues in 10% neutral buffered formalin for 24-48 hours

    • Process and embed in paraffin following standard protocols

    • Cut sections at 4-5 μm thickness

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)

    • Pressure cooker treatment for 20 minutes is often effective for FN1

  • Blocking and antibody incubation:

    • Block endogenous peroxidase with 3% H₂O₂ for 10 minutes

    • Block non-specific binding with 5-10% normal serum from the same species as the secondary antibody

    • Apply primary FN1 antibody at dilutions of 1:50-1:300 (for manual IHC) or 1:400-1:1600 (for automated systems like Leica Bond)

    • Incubate overnight at 4°C in a humidified chamber

    • Use appropriate detection systems (e.g., HRP/DAB)

  • Controls and validation:

    • Include positive control tissues with known FN1 expression (e.g., rat liver tissue has been verified for many FN1 antibodies)

    • Include negative controls by omitting primary antibody

    • Consider dual staining with markers of ECM or fibrosis for co-localization studies

  • Signal amplification for weak expression:

    • Employ tyramide signal amplification (TSA) if standard methods yield weak signals

    • Consider polymer-based detection systems for enhanced sensitivity

What steps are critical for validating the specificity of a new FN1 antibody?

Thorough validation of FN1 antibody specificity requires multiple complementary approaches:

  • Western blot validation:

    • Confirm single band or expected pattern of bands at the predicted molecular weight (262-285 kDa)

    • Include positive control lysates from cells known to express FN1 (e.g., HeLa cells)

    • Compare with established FN1 antibodies as reference standards

    • Perform peptide competition assays using the immunogen peptide to confirm specificity

  • Immunoprecipitation cross-validation:

    • Immunoprecipitate with the FN1 antibody and probe with a different FN1 antibody targeting a separate epitope

    • Verify protein identity using mass spectrometry after immunoprecipitation

  • Genetic manipulation controls:

    • Test antibody reactivity in FN1 knockdown/knockout models

    • Compare staining patterns in cell lines with varying FN1 expression levels

  • Cross-reactivity assessment:

    • Evaluate antibody performance across multiple species if cross-reactivity is claimed

    • Verify reactivity with recombinant FN1 protein

  • Application-specific validation:

    • For IHC/IF: Compare staining patterns with known FN1 distribution (extracellular matrix localization)

    • For function-blocking antibodies: Perform cell adhesion assays to confirm inhibitory activity

Why do researchers often observe unexpected bands or molecular weight variations when detecting FN1 by Western blot?

Several factors contribute to the complex banding patterns and molecular weight variations observed when detecting FN1:

  • Alternative splicing: FN1 has three regions subject to alternative splicing, potentially producing 20 different transcript variants, resulting in proteins of varying molecular weights .

  • Post-translational modifications: FN1 undergoes extensive glycosylation and other modifications that can alter its electrophoretic mobility, causing the observed molecular weight (approximately 285 kDa) to differ from the calculated weight (262-263 kDa) .

  • Proteolytic processing: FN1 can be cleaved into functional fragments by various proteases during sample preparation or in biological processes, generating lower molecular weight bands (including 69-71 kDa isoforms and shorter cleavage products) .

  • Dimer formation: Fibronectin naturally exists as dimers held together by disulfide bonds, which may not be completely reduced during sample preparation, resulting in very high molecular weight bands.

  • Cross-reactivity: Some antibodies may show weak cross-reactivity with other extracellular matrix proteins with similar domains.

To address these issues:

  • Ensure complete protein denaturation with adequate SDS and reducing agents

  • Use freshly prepared samples with protease inhibitors

  • Consider native gel electrophoresis if studying intact FN1 complexes

  • Validate bands using multiple antibodies targeting different epitopes

What are effective strategies for minimizing background in immunofluorescence studies with FN1 antibodies?

Reducing background in FN1 immunofluorescence requires addressing several potential sources of non-specific signals:

  • Optimized blocking:

    • Use 5-10% normal serum from the species of the secondary antibody

    • Add 0.1-0.3% Triton X-100 for better penetration in fixed cells

    • Consider using commercial blocking reagents specifically designed for fluorescence applications

    • Extend blocking time to 1-2 hours at room temperature or overnight at 4°C

  • Antibody dilution optimization:

    • Titrate antibody concentrations; start with manufacturer recommendations (typically 1:100-1:300 for FN1 antibodies) and adjust as needed

    • Use antibody diluent containing 1-3% BSA and 0.05-0.1% Tween-20

  • Washing procedures:

    • Increase washing steps (5-6 times) with PBS containing 0.05-0.1% Tween-20

    • Extend wash times to 5-10 minutes per wash

    • Use gentle agitation during washes

  • Sample preparation considerations:

    • Optimize fixation protocol (4% paraformaldehyde for 10-15 minutes is typically suitable)

    • Consider light autofluorescence quenching (e.g., 0.1% sodium borohydride treatment)

    • For tissue sections, treat with Sudan Black B (0.1-0.3%) to reduce autofluorescence

  • Secondary antibody selection:

    • Use highly cross-adsorbed secondary antibodies

    • Consider directly conjugated primary antibodies to eliminate secondary antibody issues

    • Include a secondary-only control to assess non-specific binding

  • Advanced countermeasures:

    • For tissue with high extracellular matrix content, pre-incubate sections with unconjugated Fab fragments

    • Use image acquisition settings that minimize autofluorescence detection

How should researchers address inconsistent results when comparing FN1 antibodies from different sources?

Inconsistent results between different FN1 antibodies can be systematically addressed through:

  • Epitope mapping comparison:

    • Identify the specific epitopes recognized by each antibody

    • Antibodies targeting different domains of FN1 may yield distinct staining patterns or detect different subsets of FN1 isoforms

    • Some antibodies like HFN 7.1 target functional domains (between PHSRN and RGD sites) , while others may target conserved regions

  • Protocol standardization:

    • Develop standardized protocols optimized for each antibody

    • Document and control variables that might affect results (fixation time, antigen retrieval method, incubation conditions)

    • Process samples in parallel when comparing antibodies

  • Reference standards usage:

    • Include the same positive control samples across experiments

    • Consider using recombinant FN1 protein as a standard

    • Compare results with published data for established antibodies

  • Cross-validation approach:

    • Employ multiple detection methods (WB, IHC, IF) with each antibody

    • Verify results with orthogonal techniques (qPCR, mass spectrometry)

    • Use genetic manipulation (siRNA knockdown) to confirm specificity

  • Quantitative comparison:

    • Develop quantitative metrics for comparing antibody performance

    • Document sensitivity, specificity, and signal-to-noise ratio for each antibody

    • Create a decision matrix for selecting the most appropriate antibody for specific applications

How can function-blocking FN1 antibodies be utilized to study cell-ECM interactions in 3D culture systems?

Function-blocking FN1 antibodies, such as HFN 7.1 which interferes with fibronectin binding to cell surface receptors , offer powerful tools for studying cell-ECM interactions in 3D culture:

  • Gradient inhibition studies:

    • Create spatial gradients of function-blocking antibodies within 3D matrices

    • Analyze directional cell migration in response to regional FN1 inhibition

    • Quantify cell morphology changes along the gradient using live cell imaging

  • Temporal regulation of cell adhesion:

    • Add function-blocking antibodies at specific timepoints to disrupt established adhesions

    • Monitor real-time changes in cell behavior using time-lapse microscopy

    • Correlate changes with activation/deactivation of downstream signaling pathways

  • Receptor specificity analysis:

    • Combine with specific integrin-blocking antibodies to dissect the contribution of different adhesion receptors

    • Map the functional domains of FN1 involved in specific cellular processes

    • Compare HFN 7.1 (which blocks the region between PHSRN and RGD sites) with antibodies targeting other domains

  • Matrix assembly inhibition:

    • Study fibrillogenesis in 3D culture by adding function-blocking antibodies during matrix formation

    • Analyze the structural consequences of impaired FN1 fibrillogenesis on other ECM components

    • Evaluate the mechanical properties of matrices formed under FN1 inhibition conditions

  • Controlled release systems:

    • Develop hydrogel or nanoparticle systems for controlled release of function-blocking antibodies

    • Create temporally defined windows of FN1 inhibition

    • Study the reversibility of cellular phenotypes after temporary FN1 function blockade

What insights can FN1 expression analysis provide in cancer research and how should such studies be designed?

FN1 expression analysis in cancer research provides multiple insights when properly designed:

  • Tumor microenvironment assessment:

    • Compare FN1 expression in tumor cells versus stromal components using dual immunostaining

    • Analyze correlation between stromal FN1 deposition and cancer progression

    • Examine co-localization with other ECM proteins to characterize matrix remodeling

  • EMT and metastasis investigation:

    • Monitor FN1 expression during epithelial-to-mesenchymal transition

    • Correlate FN1 levels with expression of EMT markers (E-cadherin, vimentin)

    • Track FN1 isoform switching during cancer progression using domain-specific antibodies

  • Therapeutic resistance mechanisms:

    • Evaluate FN1-mediated cell adhesion as a mechanism of drug resistance (cell adhesion-mediated drug resistance, CAM-DR)

    • Compare FN1 expression before and after treatment failure

    • Test combination of FN1-targeting strategies with conventional therapies

  • Experimental design considerations:

    • Use tissue microarrays for high-throughput analysis across multiple patient samples

    • Employ multi-parameter IHC/IF to correlate FN1 with other biomarkers

    • Validate findings in patient-derived xenografts and 3D organoid models

    • Complement protein-level analysis with transcriptomic data on FN1 splice variants

  • Clinical correlation approach:

    • Stratify patient cohorts based on FN1 expression patterns

    • Correlate FN1 expression with clinical outcomes (survival, recurrence, treatment response)

    • Develop standardized scoring systems for FN1 expression in clinical samples

How can researchers effectively use FN1 antibodies in stem cell differentiation and developmental biology studies?

FN1 antibodies offer valuable tools for studying stem cell biology and development:

  • Lineage specification monitoring:

    • Track temporal changes in FN1 expression during differentiation of pluripotent stem cells

    • Correlate FN1 isoform switching with key developmental transitions

    • Use function-blocking antibodies to determine critical windows when FN1-integrin interactions regulate cell fate decisions

  • Spatial patterning analysis:

    • Employ whole-mount immunostaining with FN1 antibodies to visualize ECM organization during embryonic development

    • Create high-resolution maps of FN1 distribution across developing tissues

    • Correlate FN1 gradients with morphogen distribution and cell migration patterns

  • Engineered microenvironments for directed differentiation:

    • Conjugate FN1 antibodies to defined regions of culture substrates to create patterned surfaces

    • Analyze how spatial restriction of FN1 function affects stem cell differentiation

    • Compare results with genetic knockdown approaches to validate antibody effects

  • Multi-parameter temporal analysis:

    • Combine live cell imaging with fixed-timepoint immunostaining for FN1

    • Use pulse-chase experiments with ECM labeling to track matrix turnover during differentiation

    • Correlate changes in FN1 organization with cytoskeletal remodeling and cell shape changes

  • Experimental recommendations:

    • Include careful validation of antibody specificity, as FN1 structure changes during development

    • Optimize fixation protocols to preserve ECM structure while allowing antibody penetration

    • Use super-resolution microscopy to resolve nanoscale organization of FN1 fibrils

    • Complement antibody-based studies with reporter systems for live tracking of FN1 dynamics

How should researchers quantify and interpret changes in FN1 expression patterns across different experimental conditions?

Rigorous quantification and interpretation of FN1 expression changes require systematic approaches:

  • Western blot quantification:

    • Use internal loading controls appropriate for your experimental system

    • Employ densitometry software to quantify band intensity

    • Account for potential molecular weight variants (262-285 kDa)

    • Present data as fold-change relative to control conditions

    • Perform statistical analysis across multiple biological replicates (minimum n=3)

  • Immunofluorescence quantification methods:

    • Develop standardized image acquisition parameters (exposure time, gain, offset)

    • Analyze both signal intensity and pattern (fibrillar vs. diffuse)

    • Quantify parameters such as:

      • Mean fluorescence intensity (cellular or regional)

      • Fibril length, thickness, and orientation using specialized software

      • Colocalization coefficients with cell surface markers or other ECM proteins

    • Use automated image analysis algorithms to reduce bias

  • Interpretation frameworks:

    • Consider both cellular and matrix-associated FN1 separately

    • Distinguish between changes in expression level versus altered localization

    • Account for the extracellular nature of FN1 when interpreting results

    • Remember that plasma FN1 (soluble dimeric form) is primarily secreted by hepatocytes, while cellular FN1 is produced by fibroblasts, epithelial cells, and other cell types

  • Contextual analysis:

    • Interpret FN1 changes in context of other ECM components

    • Consider the role of FN1 in your specific biological system (wound healing, development, disease)

    • Validate findings with complementary techniques (qPCR for transcript levels, proteomics)

  • Reporting standards:

    • Clearly describe quantification methods in publications

    • Include representative images alongside quantitative data

    • Report both mean values and measures of variability

    • Present raw data points alongside statistical summaries

What approaches can help distinguish between cellular FN1 production and matrix-incorporated FN1 in complex tissue samples?

Distinguishing cellular production from matrix incorporation of FN1 requires specialized techniques:

  • Differential extraction protocols:

    • Sequential extraction to separate soluble, membrane-associated, and matrix-bound FN1

    • Compare extracts using Western blotting with the same FN1 antibody

    • Quantify the distribution across different fractions

  • Cell-type specific markers in dual immunostaining:

    • Co-stain for FN1 and cell-type specific markers

    • Use nuclear counterstains to identify cellular boundaries

    • Employ confocal microscopy with z-stack analysis to distinguish intracellular from extracellular signals

    • Analyze colocalization with ER/Golgi markers to identify actively producing cells

  • In situ hybridization combined with immunostaining:

    • Detect FN1 mRNA using in situ hybridization to identify producing cells

    • Follow with FN1 immunostaining on the same section

    • Compare patterns to distinguish cells producing FN1 from areas of matrix incorporation

  • Pulse-chase experimental design:

    • Label newly synthesized proteins using click chemistry approaches

    • Track the fate of labeled FN1 from intracellular to extracellular compartments

    • Analyze incorporation into the existing matrix over time

  • Functional biochemical discrimination:

    • Use domain-specific antibodies that distinguish cellular and plasma FN1 isoforms

    • Analyze deoxycholate-soluble (newly synthesized) versus deoxycholate-insoluble (matrix-incorporated) FN1

    • Consider the relationship between fibril formation and FN1 incorporation into stable matrix structures

How can researchers accurately analyze post-translational modifications of FN1 using antibody-based approaches?

Analysis of FN1 post-translational modifications requires specialized strategies:

  • Modification-specific antibodies:

    • Use antibodies that specifically recognize phosphorylated, glycosylated, or otherwise modified FN1

    • Validate specificity using enzymatic treatment (e.g., deglycosylation, dephosphorylation)

    • Combine with pan-FN1 antibodies to determine modified fraction

  • Sequential immunoprecipitation approach:

    • First immunoprecipitate total FN1 using a general FN1 antibody

    • Then probe the immunoprecipitate with modification-specific antibodies

    • Compare ratios across experimental conditions

  • Mass spectrometry validation:

    • Immunoprecipitate FN1 and perform LC-MS/MS analysis

    • Map identified modifications to specific domains

    • Correlate antibody-based detection with MS results for validation

  • Functional correlation studies:

    • Correlate detection of specific modifications with functional assays

    • Analyze how modifications affect FN1-dependent cell behaviors

    • Use site-directed mutagenesis of modification sites to confirm functional relevance

  • Cautions and controls:

    • Include samples treated with modifying or demodifying enzymes as controls

    • Consider the effects of sample preparation on preserving labile modifications

    • Validate findings using multiple antibody clones when available

    • Remember that modifications may affect antibody accessibility to epitopes

This approach allows for detailed characterization of FN1 post-translational modifications, providing insights into how these changes affect protein function in different physiological and pathological contexts.

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.