SIGF Antibody

Shipped with Ice Packs
In Stock

Description

The SIGF polyclonal antibody is generated by immunizing a rabbit with a recombinant Arabidopsis thaliana SIGF protein segment (amino acids 296-547). Several repeated immunizations induce an immune reaction in the rabbit, resulting in the production of polyclonal antibodies against SIGF. The collected rabbit serum contains polyclonal antibodies, which are purified using affinity chromatography. The functionality of the SIGF antibody is assessed through ELISA and WB assays, verifying its ability to detect the Arabidopsis thaliana SIGF protein in experimental settings.

Product Specs

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

The SIGF polyclonal antibody is generated by immunizing a rabbit with a recombinant Arabidopsis thaliana SIGF protein segment (amino acids 296-547). Multiple repeated immunizations elicit an immune response in the rabbit, resulting in the production of polyclonal antibodies specifically targeting SIGF. The collected rabbit serum, containing these polyclonal antibodies, undergoes purification using affinity chromatography. The functionality of the SIGF antibody is rigorously assessed through ELISA and WB assays, confirming its capability to reliably detect the Arabidopsis thaliana SIGF protein in experimental settings.

Form
Liquid
Lead Time
Typically, we are able to dispatch products within 1-3 business days of receiving your order. Delivery times may vary depending on the chosen purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
RNA polymerase sigma factor sigF, chloroplastic (Sigma factor F) (Sigma-F) (Protein SINGLET OXYGEN-LINKED DEATH ACTIVATOR 8) (RNA polymerase sigma factor sig6) (Atsig6) (Sigma factor 6) SIGF SIG6 SOLDAT8 At2g36990 T1J8
Target Names
SIGF
Uniprot No.

Target Background

Function

Sigma factors are initiation factors that facilitate the binding of plastid-encoded RNA polymerase (PEP) to specific initiation sites, followed by their release. They play a crucial role in regulating transcription within chloroplasts in a DG1-dependent manner. SIGF is involved in light-dependent chloroplast development and is essential for early plant development and the formation of primary leaves.

Gene References Into Functions
  1. The interaction between DG1 and SIG6 regulates RNA polymerase-dependent chloroplast gene transcription. PMID: 20626654
  2. AtSIG6 serves as a substrate for regulatory phosphorylation by cpCK2. PMID: 20088902
  3. AtSIG6 potentially functions as a primary general sigma factor in chloroplasts during the early stages of plant development. PMID: 15807777
  4. The developmental expression of Sig6 in plastids has been reported. PMID: 16905663
  5. Inactivation of the SOLDAT8 protein does not impair oxygen singlet-mediated signaling; instead, it enhances light sensitivity. [SIGMA6] PMID: 19273469

Show More

Hide All

Database Links

KEGG: ath:AT2G36990

STRING: 3702.AT2G36990.1

UniGene: At.1453

Protein Families
Sigma-70 factor family
Subcellular Location
Plastid, chloroplast.
Tissue Specificity
Expressed in seedling, accumulating progressively. Present in leaves but not in roots.

Q&A

What is Siglec-F and how does it function in murine immunology?

Siglec-F is an innate immune receptor expressed primarily on mouse eosinophils, functioning as a critical regulator of eosinophil survival and activity. As a member of the sialic acid-binding immunoglobulin-like lectin family, Siglec-F contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) that mediates inhibitory signaling when the receptor is engaged. The functional engagement of Siglec-F leads to apoptotic pathways in eosinophils, making it an important target for studying eosinophil-mediated conditions. Research demonstrates that Siglec-F engagement triggers programmed cell death through both caspase-dependent and reactive oxygen species (ROS)-dependent mechanisms, similar to its human counterpart Siglec-8 .

How can researchers confirm successful Siglec-F antibody binding and activity?

Confirming successful Siglec-F antibody binding requires multiple validation approaches. Flow cytometry represents the primary method, allowing researchers to detect Siglec-F expression using fluorescently labeled antibodies. When evaluating antibody activity, researchers should examine markers of apoptosis such as Annexin-V positivity. In published studies, effective Siglec-F antibody engagement increased Annexin-V-positive eosinophils by approximately 76.7 ± 16.2% compared to controls within 2 hours of administration . Additionally, researchers should verify specificity by confirming that the antibody affects only Siglec-F-expressing cells (primarily eosinophils) while sparing other cell populations like mast cells. Western blotting and immunoprecipitation provide complementary approaches for verifying antibody specificity when working with cell lysates or tissue samples.

What are the key differences between in vitro and in vivo applications of Siglec-F antibodies?

In vitro and in vivo applications of Siglec-F antibodies present distinct methodological considerations. For in vitro studies, isolated eosinophils (typically from spleen or bone marrow of IL-5 transgenic mice) are directly incubated with the antibody, permitting controlled observation of direct cellular effects. Research shows that in vitro incubation with Siglec-F antibodies increases the percentage of Annexin-V-positive eosinophils from 23.5 ± 8.6% (control) to 39.9 ± 11.5% at 24 hours .

In vivo applications involve systemic administration, typically through intravenous injection at doses ranging from 20-100 μg/mouse. This approach allows assessment of physiological relevance but introduces additional variables such as antibody distribution, half-life, and potential compensatory mechanisms. In vivo studies demonstrate rapid effects, with significant eosinophil reduction observed within 2 hours post-administration and sustained reduction for at least 48 hours (84.0 ± 5.4% decrease at 2 hours and 67.0 ± 21.0% at 48 hours) . When designing either application, researchers must consider appropriate controls, including isotype-matched antibodies and binding antibodies against unrelated cell surface proteins (e.g., CD44).

Which mouse models are most appropriate for Siglec-F antibody research?

Several mouse models have proven valuable for Siglec-F antibody research, each offering unique advantages depending on the research question:

Mouse ModelKey CharacteristicsResearch ApplicationsConsiderations
IL-5 Transgenic MiceOverexpression of IL-5 leads to constitutive eosinophiliaIdeal for studying eosinophil reduction mechanismsBaseline eosinophil counts are artificially elevated
F/P+ Bone Marrow Transduced MiceModel of hypereosinophilic syndrome/chronic eosinophilic leukemia (HES/CEL)Suitable for testing therapeutic potential in eosinophilic disordersDisease progression may affect interpretation of results
Wild-type MiceNormal immune systemAppropriate for studying physiological relevanceMay require induction of eosinophilia for certain studies
Allergen-Challenged MiceInduced eosinophilic inflammationUseful for investigating allergic conditionsTiming of antibody administration relative to challenge is critical

Research indicates that Siglec-F antibody treatment is at least as efficient in reducing eosinophil levels in wild-type mice as in IL-5 transgenic mice, suggesting broad applicability across different models .

How do the mechanisms of Siglec-F antibody-induced eosinophil apoptosis compare with other eosinophil-targeting approaches?

Siglec-F antibody-induced eosinophil apoptosis represents a distinctive targeting approach compared to other eosinophil-directed therapies. The mechanism involves direct engagement of the Siglec-F receptor, triggering intracellular signaling cascades that lead to programmed cell death. This contrasts with cytokine-blocking approaches (e.g., anti-IL-5 therapies) that prevent eosinophil development and survival by interrupting growth factor signaling.

A comparative analysis reveals several mechanistic differences:

  • Siglec-F antibody induces direct apoptosis through receptor engagement, whereas IL-5 antagonists work by depleting survival factors

  • Siglec-F targeting demonstrates cell-type specificity, affecting primarily eosinophils while sparing other leukocytes, as evidenced by unaltered total white blood cell counts in treated mice

  • The apoptotic effect occurs rapidly (within 2 hours) compared to the slower effects of cytokine blockade

  • Unlike chemokine receptor antagonists that prevent eosinophil trafficking, Siglec-F antibodies actively eliminate existing eosinophil populations

The execution phase of apoptosis likely involves both caspase-dependent mechanisms and reactive oxygen species (ROS) generation, similar to what has been observed with the human ortholog Siglec-8. Researchers should consider these mechanistic distinctions when designing comparative studies or developing combination therapeutic approaches.

What are the potential confounding factors when interpreting Siglec-F antibody experimental results?

Interpreting Siglec-F antibody experimental results requires careful consideration of several potential confounding factors:

  • Fc Receptor Engagement: Antibody effects might partially result from Fc receptor-mediated mechanisms rather than direct Siglec-F signaling. Studies addressing this concern have used FcγRII/III antibody (2.4G2) pretreatment, which did not alter the observed apoptotic effects, suggesting Fc-independent mechanisms .

  • Altered Migration vs. Apoptosis: Decreased peripheral blood eosinophil counts could result from altered trafficking rather than cell death. This alternative explanation has been addressed by demonstrating increased Annexin-V positivity both in vivo and in vitro, confirming the apoptotic mechanism .

  • Variability in Siglec-F Expression: Eosinophils may exhibit heterogeneous Siglec-F expression depending on activation state and tissue localization. Researchers should characterize the expression profile of their target population before interpreting antibody effects.

  • Compensatory Mechanisms: Prolonged Siglec-F blockade may trigger compensatory production of eosinophils, potentially masking long-term efficacy. Extended time-course studies are essential for capturing these delayed responses.

  • Antibody Internalization: Treatment may induce receptor internalization, potentially complicating interpretation of whether observed effects result from signaling or from removal of functional surface receptors .

Rigorous experimental design should include appropriate controls addressing these variables, such as isotype-matched antibodies, comprehensive time-course analyses, and parallel assessment of multiple apoptosis markers.

How can computational approaches enhance antibody engineering for improved Siglec-F targeting?

Advanced computational approaches offer powerful tools for engineering antibodies with enhanced Siglec-F targeting properties. Drawing from recent advances in antibody design, researchers can employ several computational strategies:

  • Binding Mode Identification: Biophysics-informed models can identify and disentangle distinct binding modes associated with specific ligands, enabling the prediction of antibody variants with customized specificity profiles. Such approaches have successfully generated antibodies with either specific high affinity for particular targets or cross-specificity for multiple related targets .

  • Sequence-Structure-Function Modeling: Using training data from experimentally selected antibodies, researchers can develop computational frameworks that associate each potential ligand with a distinct binding mode. This enables the prediction and generation of specific variants beyond those observed in experiments .

  • Energy Function Optimization: Novel antibody sequences with predefined binding profiles can be generated by optimizing energy functions associated with each binding mode. For specific sequences, designers can minimize functions associated with desired ligands while maximizing those associated with undesired targets .

  • High-Throughput Sequencing Analysis: Integrating computational analysis with high-throughput sequencing data from phage display experiments allows researchers to identify antibody variants with desired specificity profiles, even when targeting very similar epitopes .

These computational approaches provide particular value for Siglec-F targeting by allowing researchers to design antibodies that specifically recognize Siglec-F while avoiding cross-reactivity with other Siglec family members, thereby enhancing specificity and potentially reducing off-target effects.

What are the critical parameters for flow cytometric analysis of Siglec-F antibody effects?

Designing robust flow cytometry experiments for Siglec-F antibody research requires optimization of several critical parameters:

  • Panel Design: Construct comprehensive panels that include:

    • Forward/side scatter parameters for identifying cell populations (FSC vs. SSC)

    • Dead cell discrimination dyes

    • Eosinophil markers (CCR3, Siglec-F)

    • Apoptosis markers (Annexin-V, 7-AAD)

    • Relevant activation markers

  • Instrument Selection: The choice of flow cytometer should align with experimental requirements. For standard applications with limited markers, instruments like BD FACS Canto may suffice. For more complex panels or when dealing with highly similar fluorophores, spectral cytometers like Cytek Aurora offer advantages .

  • Laser and Fluorochrome Selection: Consider the available laser configurations (UV 355nm, violet 405nm, blue 488nm, red 635nm) when selecting fluorochromes, placing bright fluorochromes on dim markers and dim fluorochromes on bright markers .

  • Gating Strategy: Implement a sequential gating approach:

    • Size/shape discrimination (FSC vs. SSC)

    • Singlet selection (Area vs. Height)

    • Dead cell exclusion

    • Leukocyte identification (CD45+)

    • Eosinophil identification (CCR3+, Siglec-F+)

    • Apoptosis assessment (Annexin-V, 7-AAD)

  • Controls: Include comprehensive controls:

    • Unstained samples

    • Single-stained compensation controls

    • Fluorescence-minus-one (FMO) controls

    • Isotype-matched antibody controls

    • Positive apoptosis controls (e.g., staurosporine-treated cells)

Researchers should also consider tissue-specific factors when analyzing Siglec-F expression and antibody effects across different compartments (blood, bone marrow, spleen, lung), as expression levels and susceptibility to antibody-induced apoptosis may vary.

How should dose-response studies for Siglec-F antibodies be designed for maximal informational value?

Designing informative dose-response studies for Siglec-F antibodies requires methodical approaches that capture both efficacy and mechanism:

  • Dose Range Selection: Establish a wide dose range spanning at least 2-3 logs, informed by literature values. Previous studies have demonstrated efficacy at 20-100 μg/mouse for in vivo administration and 1-10 μg/ml for in vitro experiments .

  • Temporal Parameters: Implement a comprehensive time-course analysis including:

    • Early timepoints (1-2 hours) to capture immediate apoptotic effects

    • Intermediate timepoints (4-24 hours) to evaluate sustained responses

    • Extended timepoints (48-72+ hours) to assess potential compensatory mechanisms

  • Readout Selection: Incorporate multiple complementary readouts:

    • Absolute eosinophil counts in blood/tissues

    • Percentage of apoptotic eosinophils (Annexin-V+)

    • Late apoptosis/necrosis markers (7-AAD+)

    • Biochemical markers of apoptotic signaling (caspase activation, mitochondrial potential)

    • Functional assessments (respiratory burst, degranulation capacity)

  • Control Antibodies: Include parallel dose-response curves for:

    • Isotype-matched control antibodies

    • Non-apoptosis-inducing binding antibodies (e.g., anti-CD44)

    • F(ab')2 fragments to assess Fc-independent effects

  • Statistical Design: Employ robust statistical approaches:

    • Calculate EC50 values with 95% confidence intervals

    • Apply appropriate regression models (four-parameter logistic for full curves)

    • Perform ANOVA with post-hoc tests for comparing multiple doses

    • Consider developing quantitative systems pharmacology models for integrating dose-response with pharmacokinetic data

These design elements collectively provide mechanistic insights beyond simple efficacy, potentially revealing threshold effects, maximal responses, and dose-limiting factors that inform both research applications and therapeutic development.

What controls are essential for validating Siglec-F antibody specificity in complex tissue environments?

Validating Siglec-F antibody specificity in complex tissue environments requires a comprehensive control strategy that addresses multiple potential confounding factors:

  • Genetic Controls:

    • Siglec-F knockout mice provide the gold standard negative control for antibody specificity

    • Selective cell-type Siglec-F deletion (using Cre-Lox systems) helps identify cell-specific effects

    • Transgenic overexpression models can serve as positive controls with enhanced signal

  • Antibody-Based Controls:

    • Isotype-matched control antibodies at equivalent concentrations

    • Pre-absorption of the antibody with recombinant Siglec-F protein

    • Competitive binding with unlabeled antibody to demonstrate specific displacement

    • F(ab')2 and Fab fragments to exclude Fc-mediated effects

    • Pre-treatment with FcγRII/III antibody (2.4G2) to block potential Fc receptor interactions

  • Cell-Type Specificity Controls:

    • Parallel assessment of Siglec-F-negative cell populations (e.g., lymphocytes)

    • Analysis of cells with low versus high Siglec-F expression

    • Comparison of effects on multiple potential Siglec-F-expressing cell types

  • Technical Validation:

    • Multiple antibody clones targeting different Siglec-F epitopes

    • Secondary antibody-only controls

    • Fluorescence-minus-one (FMO) controls for flow cytometry

    • Blocking peptides for immunohistochemistry applications

  • Cross-Reactivity Assessment:

    • Testing against related Siglec family members

    • Heterologous expression systems expressing individual Siglec proteins

When working with tissues known to have high autofluorescence (e.g., lung), researchers should implement additional controls including unstained samples and spectral unmixing approaches . The combined use of these controls provides robust validation of antibody specificity even in complex tissue environments with multiple cell types and potential interfering factors.

How should researchers address variability in Siglec-F antibody responses across different experimental models?

Addressing variability in Siglec-F antibody responses requires systematic analytical approaches that account for model-specific factors:

  • Standardization of Response Metrics:

    • Calculate relative changes (percent decrease from baseline) rather than absolute values

    • Develop normalized response indices that incorporate multiple parameters

    • Establish internal reference standards for each experimental batch

  • Model-Specific Analysis:

    • In IL-5 transgenic models, account for baseline hyperesosinophilia when interpreting magnitude of response

    • For disease models (e.g., F/P+ HES/CEL model), stratify analysis based on disease severity

    • In allergen challenge models, normalize responses to the degree of initial inflammatory response

  • Covariates and Confounders:

    • Document and account for age, sex, genetic background, and microbiome variability

    • Implement multivariate analysis to identify factors significantly influencing response

    • Consider mixed-effects models that incorporate both fixed and random effects

  • Meta-analytical Approaches:

    • Pool data across multiple experiments using standardized effect sizes

    • Implement random-effects meta-analysis when heterogeneity is present

    • Conduct sensitivity analyses to identify influential outliers

  • Response Heterogeneity Analysis:

    • Characterize responder vs. non-responder phenotypes

    • Implement cluster analysis to identify response patterns

    • Investigate potential mechanisms underlying differential responses

What are the limitations of current methodologies for detecting Siglec-F antibody-induced apoptosis in tissue-resident eosinophils?

Current methodologies for detecting Siglec-F antibody-induced apoptosis in tissue-resident eosinophils face several important limitations that researchers must address:

  • Rapid Clearance Challenges: Apoptotic eosinophils in tissues are rapidly cleared by resident macrophages, potentially leading to underestimation of apoptotic events. Unlike circulating eosinophils, where apoptotic changes can be readily captured (with an observed 76.7 ± 16.2% increase in Annexin-V positivity within 2 hours of antibody administration) , tissue-resident cells may be removed before detection.

  • Tissue Processing Artifacts: Standard tissue digestion protocols may selectively lose apoptotic cells or induce artifactual apoptosis, complicating interpretation. Anexin-V staining is particularly susceptible to calcium-chelating agents commonly used in tissue processing.

  • Phenotypic Heterogeneity: Tissue-resident eosinophils display phenotypic heterogeneity with variable Siglec-F expression levels across different tissue compartments, potentially affecting susceptibility to antibody-induced apoptosis.

  • Microenvironmental Influences: Local tissue factors (cytokines, adhesion molecules, extracellular matrix) may modulate Siglec-F signaling and apoptotic responses, creating context-dependent effects not observed in blood or isolated cells.

  • Technical Detection Limitations:

    • Autofluorescence in certain tissues (particularly lung) interferes with flow cytometric detection

    • Immunohistochemical approaches lack the sensitivity to distinguish early apoptotic events

    • In situ markers of apoptosis (TUNEL, cleaved caspase-3) may underrepresent the total apoptotic population

To overcome these limitations, researchers should implement complementary approaches, including real-time intravital microscopy, macrophage depletion strategies to prevent clearance, and tissue-specific flow cytometry panels optimized for high autofluorescence environments using spectral analyzers like the Cytek Aurora . Additionally, mathematical modeling that accounts for apoptotic cell clearance kinetics can help estimate the true rate of antibody-induced apoptosis even when direct detection is challenging.

How can researchers integrate Siglec-F antibody findings with broader immunological contexts for translational relevance?

Integrating Siglec-F antibody research into broader immunological and translational contexts requires sophisticated analytical frameworks that connect mechanistic insights to potential clinical applications:

  • Cross-Species Translation:

    • Conduct comparative analyses between Siglec-F (mouse) and Siglec-8 (human) mechanisms

    • Develop parallel experimental systems in human samples and murine models

    • Create humanized mouse models expressing human Siglec-8 for translational studies

    • Apply biophysics-informed computational models to predict cross-species antibody specificity profiles

  • Systems Immunology Approaches:

    • Implement multi-parameter analyses to position Siglec-F pathways within broader immune networks

    • Conduct transcriptomic profiling before and after antibody treatment to identify affected pathways

    • Apply computational modeling to predict consequences of eosinophil depletion on other immune compartments

    • Integrate findings with other eosinophil-regulatory mechanisms

  • Disease-Specific Contextualization:

    • Compare Siglec-F antibody efficacy across multiple disease models (allergic, parasitic, hypereosinophilic)

    • Conduct correlative analyses between treatment response and disease-specific biomarkers

    • Evaluate Siglec-F targeting in conjunction with standard-of-care therapies for relevant conditions

    • Assess differential effects on inflammatory versus homeostatic eosinophil populations

  • Biomarker Integration:

    • Develop predictive biomarkers of response to Siglec-F targeting

    • Establish pharmacodynamic markers that reflect successful target engagement

    • Correlate Siglec-F expression patterns with disease phenotypes

    • Identify potential companion diagnostics for translated therapies

  • Therapeutic Paradigm Development:

    • Position Siglec-F targeting within existing treatment algorithms for eosinophilic disorders

    • Conduct comparative analyses with other eosinophil-depleting approaches (e.g., anti-IL-5)

    • Explore combination strategies targeting multiple eosinophil regulatory pathways

    • Develop precision medicine approaches for patient stratification

This integrative approach enhances translational relevance by contextualizing the mechanistic finding that Siglec-F antibody administration selectively reduces blood and tissue eosinophils within broader therapeutic paradigms for eosinophilic disorders such as asthma, hypereosinophilic syndromes, and eosinophilic gastrointestinal diseases.

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.