ANXA1 Mouse

Annexin A1 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to ANXA1 Mouse

The "ANXA1 Mouse" refers to a genetically engineered Annexin A1 (ANXA1) knockout (KO) mouse model, generated by disrupting the Anxa1 gene via homologous recombination. This model is pivotal for studying ANXA1's roles in inflammation, angiogenesis, metabolism, and cancer biology. ANXA1 is a glucocorticoid-regulated protein with calcium-dependent phospholipid-binding properties, implicated in resolving inflammation, immune modulation, and vascular remodeling .

Role in Angiogenesis and Tumor Biology

ANXA1-KO mice exhibit profound defects in pathological angiogenesis and tumor progression:

Key Findings:

  • Impaired tumor growth: Tumors in ANXA1-KO mice grow 33% slower than in wild-type (WT) mice, with reduced metastasis and increased necrosis .

  • Defective endothelial sprouting: Aortic rings from KO mice show 50% fewer vascular sprouts, rescued by ANXA1 re-expression .

  • Altered gene expression: Upregulation of anti-angiogenic TIMP2 and downregulation of pro-angiogenic SphK1 in tumors .

Table 1: Tumor and Angiogenesis Phenotypes in ANXA1-KO Mice

ParameterANXA1-KO vs. WTCitation
Tumor vessel density↓ 66%
Metastasis incidence↓ 70%
Wound healing rate↓ 40%

Cardiovascular Implications

ANXA1 deficiency exacerbates cardiac injury and inflammation post-myocardial infarction (MI):

  • Increased infarct size: 25% larger infarcts in KO mice after ischemia-reperfusion .

  • Elevated inflammation: Upregulated CD68 (macrophages), CCR2 (monocytes), and NLRP3 (inflammasome) in KO hearts .

  • Hematopoietic dysregulation: Enhanced HSPC mobilization and neutrophilia 8 days post-MI .

Immune System Regulation

ANXA1-KO mice display hyperinflammatory responses:

  • Leukocyte migration: Increased neutrophil infiltration in stroke and colitis models .

  • Cytokine imbalance: Elevated pro-inflammatory cytokines (e.g., IL-1β) in inflamed tissues .

  • Therapeutic rescue: Exogenous ANXA1 administration reverses inflammatory phenotypes .

Metabolic and Adipose Tissue Effects

Female ANXA1-KO mice on high-fat diets (HFD) develop metabolic dysfunction:

  • Adiposity: 20% higher body weight and fat mass vs. WT .

  • Insulin resistance: Fasting glucose ↑ 30%, insulin ↑ 50% in KO mice .

  • Lipolytic defects: Impaired upregulation of ATGL and HSL enzymes in adipose tissue .

Platelet Function and Haemostasis

ANXA1 modulates platelet activation without affecting baseline haemostasis:

  • Receptor upregulation: Platelets from KO mice show 2-fold higher Fpr2/3 expression .

  • Activation threshold: Reduced response to low-dose agonists (e.g., collagen), but normal tail-bleeding times .

Therapeutic Implications

Targeting ANXA1 shows promise in cancer therapy:

  • MDX-124 antibody: Reduces proliferation in ANXA1-expressing cancers (e.g., triple-negative breast cancer) by inducing G1 cell-cycle arrest .

  • Preclinical efficacy: 60% tumor growth inhibition in syngeneic mouse models .

Product Specs

Introduction
ANXA1, a member of the Ca(2+)-dependent phospholipid binding protein family, is localized to the cytosolic face of the plasma membrane and has a molecular weight ranging from 35kDa to 40kDa. With a molecular weight of 40kDa, ANXA1 exhibits high-affinity binding to two to four calcium ions and possesses phospholipase A2 inhibitory activity. Given the role of phospholipase A2 in the biosynthesis of prostaglandins and leukotrienes, potent inflammatory mediators, ANXA1 is thought to have potential anti-inflammatory effects. Moreover, ANXA1 plays a crucial role in exocytosis by promoting membrane fusion. Immunocytochemical recognition of ANXA1 protein provides a straightforward, highly sensitive, and specific assay for diagnosing hairy cell leukemia.
Description
Recombinant ANXA1 Mouse, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 369 amino acids (1-346 a.a), has a molecular mass of 41.1 kDa, and is fused to a 23 amino acid His-tag at its N-terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The ANXA1 protein solution (1mg/ml) is supplied in a buffer consisting of phosphate buffered saline (pH 7.4), 20% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the product frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for extended storage periods. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 95.0% using SDS-PAGE analysis.
Synonyms
Annexin A1, Annexin I, Annexin-1, Calpactin II, Calpactin-2, Chromobindin-9, Lipocortin I, Phospholipase A2 inhibitory protein, p35.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMAMVSEF LKQARFLENQ EQEYVQAVKS YKGGPGSAVS PYPSFNVSSD VAALHKAIMV KGVDEATIID ILTKRTNAQR QQIKAAYLQE NGKPLDEVLR KALTGHLEEV VLAMLKTPAQ FDADELRGAM KGLGTDEDTL IEILTTRSNE QIREINRVYR EELKRDLAKD ITSDTSGDFR KALLALAKGD RCQDLSVNQD LADTDARALY EAGERRKGTD VNVFTTILTS RSFPHLRRVF QNYGKYSQHD MNKALDLELK GDIEKCLTTI VKCATSTPAF FAEKLYEAMK GAGTRHKALI RIMVSRSEID MNEIKVFYQK KYGISLCQAI LDETKGDYEK ILVALCGGN.

Q&A

What is ANXA1 and what cellular functions does it regulate in mice?

ANXA1 (Annexin A1) is a calcium-dependent phospholipid binding protein that functions as a key regulator of inflammatory responses. Initially characterized as an anti-phospholipase A2 and glucocorticoid-inducible 37-kDa protein, ANXA1 plays critical roles in inflammation, leukocyte migration and accumulation, phagocytosis, cell signaling, apoptosis, and membrane trafficking . In mouse models, ANXA1 has demonstrated regulatory functions in inflammatory cell adhesion to endothelial cells, with endogenous ANXA1 becoming externalized to the cell surface during the early stages of inflammation, where it exerts antiadhesive properties to modulate polymorphonuclear leukocyte (PMN) transmigration .

What ANXA1 mouse models are available for research?

The primary mouse models for ANXA1 research include:

  • ANXA1 knockout (ANXA1-/-): Complete genetic deletion of ANXA1, created through various targeting strategies including the MGI:4363865 Anxa1<tm1a(KOMP)Wtsi> allele .

  • Conditional knockout models: Tissue-specific deletion of ANXA1 using Cre-loxP technology.

  • ANXA1 overexpression models: Transgenic mice overexpressing wild-type or modified ANXA1.

  • ANXA1 point mutation models: Mice harboring specific mutations in ANXA1 protein sequence.

When selecting a model, researchers should consider the specific research question, experimental timeline, and desired readouts. Characterization of baseline phenotypes is essential before proceeding with experimental interventions.

How do I confirm ANXA1 deletion in knockout mice?

Verification of ANXA1 deletion should employ multiple complementary approaches:

  • Genotyping PCR: Using primers specific to the wildtype and modified alleles.

  • Protein expression analysis: Western blotting of tissue lysates using ANXA1-specific antibodies.

  • mRNA expression: qRT-PCR to confirm absence of ANXA1 transcripts.

  • Immunohistochemistry: To verify tissue-specific absence of ANXA1 protein.

  • Functional assays: Assessing known ANXA1-dependent processes.

What are the key considerations when designing myocardial infarction experiments using ANXA1 knockout mice?

When designing myocardial infarction (MI) studies with ANXA1-/- mice, researchers should consider:

  • Appropriate timepoints: For assessment of cardiac necrosis and early cardiac inflammation, 24h and 48h post-MI are optimal timepoints . For HSPC mobilization and regional inflammation assessment, 8 days post-MI is recommended. For long-term cardiac physiology changes, 4-week timepoints are appropriate .

  • Surgical protocol standardization: Consistent coronary artery occlusion technique is critical for reproducible infarct size.

  • Control groups: Include both sham-operated ANXA1-/- mice and wildtype counterparts with matched procedures.

  • Comprehensive analysis: Infarct size measurement, cardiac function (echocardiography), inflammatory cell profiling (flow cytometry), and gene expression analysis should be performed to capture the multifaceted effects of ANXA1 deficiency.

  • Compensatory mechanism assessment: Evaluate expression of related proteins (FPR1, FPR2) that may be upregulated as compensatory mechanisms .

In previous studies, ANXA1-/- mice showed exaggerated inflammatory responses with significantly higher macrophage density and infarct size compared to WT mice after MI , emphasizing the importance of monitoring both cardiac damage and inflammatory parameters.

How should I design experiments to investigate ANXA1's role in angiogenesis and wound healing?

For angiogenesis and wound healing studies with ANXA1 mouse models:

  • In vitro approaches:

    • Aortic ring assays: Extract aortic rings from WT and ANXA1-/- mice, embed in Matrigel, and quantify endothelial sprouting over 7-10 days .

    • Endothelial cell isolation and culture: Compare proliferation, migration, and tube formation between genotypes.

  • In vivo approaches:

    • Wound healing models: Create standardized cutaneous wounds and monitor closure rates, tissue revascularization, and inflammatory cell infiltration.

    • Matrigel plug assays: Subcutaneous implantation of growth factor-containing Matrigel to assess in vivo angiogenesis.

    • Tumor angiogenesis models: Implant syngeneic tumor cells and analyze tumor vascular development.

  • Molecular analyses:

    • Gene expression profiling for pro- and anti-angiogenic factors

    • Immunohistochemistry for vessel density and maturation markers

    • Flow cytometry for quantification of endothelial progenitor cells

Previous research demonstrated that aortic rings from ANXA1-KO mice exhibit impaired endothelial cell sprouting that can be rescued by adenoviral expression of ANXA1 . Additionally, both tumor vascular development and wound healing were significantly impaired in ANXA1-KO tissues .

How does ANXA1 deficiency affect cardiac remodeling after myocardial infarction?

ANXA1 deficiency significantly impacts cardiac remodeling post-MI through several mechanisms:

  • Exacerbated initial damage: ANXA1-/- mice exhibit increased infarct size following ischemia-reperfusion injury compared to WT mice . This establishes a more extensive baseline injury that influences subsequent remodeling.

  • Enhanced inflammatory response: ANXA1-/- hearts show significantly upregulated expression of inflammatory markers including IL-1β, TNF-α, and NLRP3 (the regulator of IL-1β maturation) . Macrophage markers CD68, CD11c, and monocyte marker CCR2 are markedly elevated (>50-fold increase for CCR2) .

  • Altered inflammatory cell profile: Hearts from ANXA1-/- mice show evidence of enhanced M1 macrophage polarization after MI, with a tendency for increased expression of S100A9 .

  • Accelerated pathological remodeling: ANXA1-/- mice develop increased heart and lung weight after MI, along with upregulated hypertrophic gene expression (8-fold increase in ANP) . These changes are early indicators of progression toward cardiac failure.

  • Hematopoietic changes: ANXA1-/- mice exhibit greater expansion of hematopoietic stem progenitor cells (HSPCs) and altered patterns of HSPC mobilization 8 days post-MI .

These findings suggest that endogenous ANXA1 plays a protective role in limiting excessive inflammation and pathological remodeling after cardiac injury. Therapeutic strategies targeting the ANXA1 pathway might therefore offer potential for mitigating adverse cardiac remodeling.

What methods should I use to analyze HSPC mobilization in ANXA1-/- mice after myocardial injury?

To comprehensively analyze HSPC mobilization in ANXA1-/- mice following myocardial injury:

  • Multicompartment sampling: Collect bone marrow, peripheral blood, and spleen to track HSPC migration between compartments. The assessment of all three compartments is critical as ANXA1-/- mice show altered distribution patterns post-MI .

  • Flow cytometry panels:

    • HSPC identification: Use markers including Lin-/Sca-1+/c-Kit+ (LSK) cells

    • Progenitor subpopulations: Identify common myeloid progenitors (CMPs), granulocyte-macrophage progenitors (GMPs), and megakaryocyte-erythroid progenitors (MEPs)

    • Mature myeloid cells: Quantify neutrophils, monocytes, and platelets

  • Colony-forming assays: Perform to assess functional capacity of mobilized HSPCs from different compartments.

  • Temporal analysis: Collect samples at multiple timepoints (baseline, 3 days, 8 days, and 14 days post-MI) to capture dynamic changes in HSPC mobilization.

  • Mobilization signaling: Measure plasma levels of HSPC mobilization factors (G-CSF, SDF-1) and their receptors.

Research has shown that ANXA1-/- mice exhibit expansion of HSPCs in bone marrow post-MI, with reduction in CMPs and MEPs, but expansion of GMPs, suggesting prioritization of neutrophil production . This is accompanied by increased circulating neutrophils and platelets, with a trend toward lower blood monocytes potentially reflecting increased myocardial infiltration .

How does host ANXA1 deficiency influence tumor growth and metastasis in mouse models?

Host ANXA1 deficiency profoundly affects tumor development through several mechanisms:

  • Reduced tumor growth: Tumors growing in ANXA1-KO mice show significantly decreased growth rates compared to those in wildtype hosts . This appears to be mediated through stromal rather than tumor cell effects.

  • Decreased metastasis: Metastatic spread is significantly reduced in ANXA1-KO mice, suggesting a role for host ANXA1 in facilitating tumor cell dissemination .

  • Increased tumor necrosis: Tumors in ANXA1-KO mice exhibit greater necrotic areas, potentially reflecting compromised tumor vasculature .

  • Improved survival: ANXA1-KO mice bearing tumors show significantly prolonged survival compared to wildtype counterparts .

  • Impaired tumor angiogenesis: Vascular development within tumors is markedly reduced in ANXA1-KO mice, likely contributing to the reduced growth and increased necrosis .

These findings highlight the critical role of host-derived ANXA1 in supporting tumor progression, particularly through pro-angiogenic functions. Systems analysis of gene expression in these tumors specifically implicates angiogenesis and wound healing in this impairment . The research suggests that targeting ANXA1 in the tumor microenvironment might represent a novel therapeutic approach for cancer treatment.

What experimental approaches can distinguish between tumor cell-intrinsic versus stromal effects of ANXA1?

To delineate tumor cell-intrinsic versus stromal effects of ANXA1, implement these complementary approaches:

  • Reciprocal transplantation studies:

    • Inject ANXA1-knockdown tumor cells into WT and ANXA1-/- mice

    • Inject ANXA1-overexpressing tumor cells into WT and ANXA1-/- mice

    • Compare growth patterns to determine relative contributions

  • Co-culture systems:

    • Establish in vitro co-cultures of tumor cells with stromal components (fibroblasts, endothelial cells) from WT or ANXA1-/- mice

    • Measure proliferation, invasion, and angiogenic factor production

  • Bone marrow chimeras:

    • Generate mice with ANXA1-/- bone marrow in WT bodies and vice versa

    • Implant tumor cells to determine contribution of bone marrow-derived versus tissue-resident ANXA1

  • Tissue-specific conditional knockouts:

    • Use conditional ANXA1 knockout in specific stromal compartments (e.g., endothelial cells, fibroblasts, or myeloid cells)

    • Analyze tumor growth and metastasis in each model

  • Molecular profiling:

    • Perform single-cell RNA sequencing of tumors grown in WT versus ANXA1-/- mice

    • Identify cell type-specific transcriptional changes

Previous research demonstrated that syngeneic tumor cells injected into ANXA1-KO mice showed impaired growth and metastasis, supporting a critical role for host-derived ANXA1 in tumor progression . This approach can be extended by manipulating ANXA1 expression in specific cellular compartments to further dissect its functions.

How can I effectively study ANXA1's role in leukocyte-endothelial interactions using intravital microscopy?

To optimize intravital microscopy studies of ANXA1's role in leukocyte-endothelial interactions:

  • Experimental setup:

    • Mouse preparation: Use WT and ANXA1-/- mice with surgically exposed microcirculation (cremaster muscle, mesentery, or ear skin)

    • Inflammatory stimulus: Apply IL-1β (10 ng) locally to induce inflammation

    • Intervention timing: For recombinant protein studies, administer AnxA1 or modified variants (e.g., SAnxA1) after establishing baseline inflammation

  • Key parameters to measure:

    • Cell flux: Quantify the number of leukocytes passing through a vessel segment per unit time

    • Rolling velocity: Measure at multiple timepoints (significant changes observed ≥10 minutes after treatment)

    • Cell adhesion: Count firmly adherent cells per vessel area

    • Transmigration: Track cells crossing the endothelial barrier

  • Pharmacological approaches:

    • Dose-response studies: Test multiple concentrations of AnxA1 or variants

    • Receptor antagonism: Use FPR2 antagonists or FPR2-/- mice to confirm mechanism specificity

    • PR3 inhibitors: Apply to prevent AnxA1 cleavage

  • Advanced analyses:

    • Real-time calcium imaging: To monitor cell activation

    • Multi-channel fluorescence: Label different cell populations and track interactions

    • Long-term imaging: Capture the temporal dynamics of AnxA1 effects

Previous research demonstrated that both AnxA1 and cleavage-resistant SAnxA1 modified ongoing inflammation by increasing cell velocity and reducing adhesion, with SAnxA1 showing more persistent effects (approximately 50% inhibition of adhesion at 30 minutes post-administration compared to 30% for AnxA1) . Importantly, these effects were abolished in mice lacking FPR2, confirming receptor specificity .

What are the recommended approaches for investigating ANXA1 cleavage by proteases in inflammatory conditions?

To investigate ANXA1 cleavage by proteases during inflammation:

  • In vitro cleavage assays:

    • Recombinant protein approaches: Incubate purified AnxA1 with increasing concentrations of recombinant PR3 (or other proteases)

    • Detection methods: Western blotting with antibodies specific to N-terminal or C-terminal regions

    • Mass spectrometry: For precise identification of cleavage sites

  • Engineered cleavage-resistant variants:

    • Site-directed mutagenesis: Modify protease recognition sites (e.g., A11R, V22K, V36K mutations for PR3 resistance)

    • Validation: Test resistance to cleavage using in vitro protease assays

  • Cellular models:

    • PMN-endothelial co-cultures: Activate neutrophils to release proteases and monitor AnxA1 cleavage

    • Confocal microscopy: Visualize membrane-bound versus cleaved AnxA1 using domain-specific antibodies

  • In vivo approaches:

    • Inflammation models: Analyze AnxA1 fragments in inflammatory exudates

    • Comparative studies: Test native AnxA1 versus cleavage-resistant variants (e.g., SAnxA1)

    • Protease inhibitors: Administer specific inhibitors to prevent AnxA1 cleavage in vivo

Research has demonstrated that systematic mutation of PR3 cleavage sites in AnxA1 (A11R/V22K/V36K triple mutant, termed SAnxA1) confers complete resistance to PR3 cleavage while maintaining biological activity . This cleavage-resistant variant shows stronger and longer-lasting anti-inflammatory effects compared to native AnxA1, suggesting that AnxA1 cleavage represents a catabolic rather than activating event .

How can I reconcile the seemingly contradictory roles of ANXA1 in different disease models?

Reconciling ANXA1's diverse and sometimes contradictory roles requires multifaceted approaches:

  • Context-dependent analysis:

    • Cell type specificity: Determine if ANXA1 functions differently in various cell types (e.g., endothelial cells versus leukocytes)

    • Disease stage discrimination: Examine acute versus chronic phases of disease models

    • Local versus systemic effects: Distinguish between tissue-specific and systemic consequences of ANXA1 modulation

  • Receptor interaction profiling:

    • Map ANXA1 interactions with different receptors (FPR1, FPR2, others) across tissues

    • Determine if receptor expression patterns explain differential outcomes

  • Post-translational modification analysis:

    • Investigate how phosphorylation, proteolytic processing, or other modifications alter ANXA1 function

    • The PR3-resistant SAnxA1 studies demonstrate how preventing proteolysis enhances anti-inflammatory effects

  • Integrated multi-omics approaches:

    • Combine transcriptomics, proteomics, and metabolomics across models

    • Identify divergent downstream pathways activated in different contexts

  • Temporal dynamics:

    • Track ANXA1 expression, localization, and function over disease progression

    • ANXA1 expression increases after ischemia-reperfusion or myocardial infarction, suggesting dynamic regulation

The apparent contradiction between ANXA1's anti-inflammatory effects in cardiovascular models and its pro-tumorigenic effects in cancer models might be explained by its fundamental role in regulating angiogenesis - beneficial for tissue repair but detrimental in tumor growth. Additionally, the timing of ANXA1 activity and its cellular source appear critical in determining outcomes across disease models.

What computational approaches can help predict novel functions of ANXA1 based on existing mouse model data?

Advanced computational approaches for predicting novel ANXA1 functions:

  • Network analysis of multi-omics data:

    • Integrate transcriptomic, proteomic, and metabolomic data from ANXA1-/- versus WT mice

    • Apply weighted gene co-expression network analysis (WGCNA) to identify modules of co-regulated genes

    • Search for novel pathways connected to ANXA1

  • Pathway enrichment and systems biology:

    • Perform gene set enrichment analysis (GSEA) across multiple ANXA1 datasets

    • Apply ingenuity pathway analysis (IPA) to predict upstream regulators and downstream effects

    • Previous systems analysis implicated specific vascular functions in ANXA1-mediated phenotypes

  • Protein-protein interaction prediction:

    • Use structural modeling to predict ANXA1 interactions beyond known partners

    • Apply machine learning algorithms to predict functional consequences of these interactions

  • Pharmacological network analysis:

    • Compare transcriptional signatures of ANXA1 modulation with drug-induced signatures

    • Identify compounds that might mimic or antagonize ANXA1 functions

  • Cross-species conservation analysis:

    • Examine evolutionary conservation of ANXA1 functions across model organisms

    • Identify highly conserved pathways likely to represent core ANXA1 functions

  • Text mining and literature-based discovery:

    • Apply natural language processing to extract hidden connections in the literature

    • Generate testable hypotheses about novel ANXA1 functions

Implementing these computational approaches could help reconcile apparently contradictory findings and identify previously unrecognized functions of ANXA1, potentially leading to novel therapeutic applications in inflammation, cardiovascular disease, and cancer.

What are the critical quality control steps for recombinant ANXA1 protein preparation for in vivo studies?

For reliable recombinant ANXA1 protein preparation for in vivo studies:

  • Expression system selection:

    • Mammalian expression: Preferred for proper post-translational modifications

    • E. coli systems: Higher yield but may lack critical modifications

    • HEK293 cells: Successfully used for MFAnxA1 mutants with double-tagging (c-Myc on N-terminus, Flag on C-terminus)

  • Purification quality control:

    • Multiple chromatography steps: Affinity chromatography followed by size exclusion

    • Endotoxin testing: Critical for in vivo applications (<0.1 EU/mg protein)

    • SDS-PAGE and Western blotting: Verify size, purity, and immunoreactivity

    • Mass spectrometry: Confirm protein identity and detect modifications

  • Functional validation:

    • Calcium and phospholipid binding assays: Confirm core annexin properties

    • Receptor binding assays: Verify FPR2 activation capacity

    • Cell-based bioactivity: Test anti-inflammatory effects on neutrophils or endothelial cells

  • Stability assessment:

    • Thermal stability testing: Using differential scanning fluorimetry

    • Storage condition optimization: Test various buffers, pH values, and additives

    • Freeze-thaw stability: Determine maximum allowable cycles

  • Pre-in vivo validation:

    • Protease resistance testing: Especially for engineered variants like SAnxA1

    • Pharmacokinetic pilot studies: Determine half-life and biodistribution

    • Dose-finding studies: Establish effective concentration range

These quality control measures are essential for ensuring that observed biological effects are attributable to the protein of interest rather than contaminants or degraded products. Research has demonstrated that properly prepared recombinant AnxA1 and SAnxA1 retain their ability to bind and activate FPR2 receptors and exhibit expected anti-inflammatory activities in vivo .

What is the optimal experimental design for studying ANXA1's impact on hematopoietic stem cell mobilization and function?

To optimally investigate ANXA1's role in hematopoietic stem cell biology:

  • Baseline characterization:

    • Quantitative analysis: Compare HSPC numbers in bone marrow, blood, and spleen of WT versus ANXA1-/- mice under steady-state conditions

    • Phenotypic profiling: Assess HSPC subpopulations (LT-HSC, ST-HSC, MPP) and committed progenitors (CMP, GMP, MEP)

    • Cell cycle analysis: Determine proliferation status and quiescence of HSPCs

    • ANXA1 expression mapping: Characterize ANXA1 expression across HSPC populations

  • Mobilization studies:

    • Inflammatory mobilization: Apply MI model or other inflammatory stimuli and track HSPC mobilization over time

    • Pharmacological mobilization: Compare G-CSF or AMD3100-induced mobilization between genotypes

    • Competitive mobilization: Co-house WT and ANXA1-/- mice connected by parabiosis to assess cell-intrinsic versus environmental factors

  • Functional assessment:

    • Colony-forming assays: Compare CFU-GM, BFU-E, and CFU-GEMM formation

    • Transplantation studies: Competitive repopulation assays with WT versus ANXA1-/- bone marrow

    • Serial transplantation: Assess long-term self-renewal capacity

  • Molecular analyses:

    • Transcriptome profiling: RNA-seq of sorted HSPCs from both genotypes

    • HSPC niche interaction: Analyze adhesion molecule expression and signaling

    • Stress response: Evaluate resistance to genotoxic and oxidative stress

Research has demonstrated that ANXA1-/- mice exhibit expansion of HSPCs in bone marrow after MI, accompanied by reduction in CMPs and MEPs but expansion of GMPs . This altered progenitor cell distribution was associated with increased circulating neutrophils and platelets, suggesting that ANXA1 regulates HSPC mobilization and lineage specification during inflammatory responses .

What emerging technologies could advance our understanding of ANXA1 biology in mouse models?

Cutting-edge technologies to enhance ANXA1 research:

  • CRISPR-based approaches:

    • Base editing: Introduce specific point mutations to study structure-function relationships

    • Prime editing: Create precise modifications without double-strand breaks

    • CRISPRa/CRISPRi: Modulate ANXA1 expression without permanent genetic changes

    • CRISPR screening: Identify genetic modifiers of ANXA1 function

  • Single-cell technologies:

    • scRNA-seq: Profile transcriptional heterogeneity in ANXA1-expressing cells

    • Spatial transcriptomics: Map ANXA1 expression patterns within tissue architecture

    • CyTOF: Analyze ANXA1 protein levels alongside dozens of other markers

  • Advanced imaging:

    • Intravital multiphoton microscopy: Track ANXA1+ cells in living tissues over time

    • Super-resolution microscopy: Visualize subcellular ANXA1 localization

    • FRET/FLIM: Monitor ANXA1-protein interactions in real-time

  • Biomaterial and organoid approaches:

    • 3D tissue models: Study ANXA1 in physiologically relevant microenvironments

    • Engineered vascular networks: Investigate ANXA1's role in angiogenesis

    • Organ-on-chip: Model complex tissue interactions under controlled conditions

  • Proteomics and interactomics:

    • Proximity labeling: Identify ANXA1 interaction partners in living cells

    • Post-translational modification mapping: Characterize regulatory modifications

    • Thermal proteome profiling: Discover novel ANXA1-binding proteins

These technologies could help resolve outstanding questions about ANXA1's seemingly contradictory roles in different disease contexts and potentially identify new therapeutic approaches targeting the ANXA1 pathway.

How might therapeutic targeting of the ANXA1 pathway be developed based on mouse model findings?

Translating ANXA1 mouse model findings toward therapeutic applications:

  • Pathway-specific intervention strategies:

    • Stable ANXA1 mimetics: Development of protease-resistant ANXA1 variants (like SAnxA1)

    • Tissue-targeted delivery: Nanoparticle formulations to direct ANXA1 or mimetics to specific tissues

    • Cell-specific activation: Technologies to induce ANXA1 expression in defined cell populations

  • Disease-specific approaches:

    • Cardiovascular applications: ANXA1 mimetics to reduce inflammation and promote healing post-MI

    • Cancer applications: ANXA1 antagonists targeting tumor angiogenesis

    • Inflammatory conditions: Stable ANXA1 variants with enhanced anti-inflammatory properties

  • Combination therapies:

    • ANXA1 + standard of care: Evaluate synergistic effects

    • Sequential therapeutic approaches: Timed administration based on disease stage

    • Multi-target strategies: Combining ANXA1 pathway modulation with complementary approaches

  • Biomarker development:

    • ANXA1 cleavage products: As indicators of inflammatory activity

    • ANXA1/FPR pathway activation: To identify responder populations

    • Pharmacodynamic markers: To optimize dosing and timing

  • Precision medicine applications:

    • Genotype-guided therapy: Based on ANXA1 pathway polymorphisms

    • Disease endotype identification: Stratify patients based on ANXA1 pathway status

    • Response prediction: Develop models to predict efficacy of ANXA1-targeted interventions

Product Science Overview

Biological Role and Functions

ANXA1 plays a crucial role in the regulation of inflammation and the immune response. It mediates some of the anti-inflammatory effects of glucocorticoids, which are hormones that modulate inflammation and immune responses . The protein is involved in various cellular processes, including:

  • Inhibition of Phospholipase A2: ANXA1 inhibits phospholipase A2, an enzyme that releases arachidonic acid from membrane phospholipids, leading to the production of pro-inflammatory eicosanoids .
  • Regulation of Leukocyte Migration: ANXA1 helps regulate the migration of leukocytes (white blood cells) to sites of inflammation, thereby controlling the extent of the inflammatory response .
  • Promotion of Apoptosis: ANXA1 promotes apoptosis (programmed cell death) in certain cell types, which is essential for the resolution of inflammation and the maintenance of tissue homeostasis .
Therapeutic Potential

Given its significant role in inflammation, ANXA1 has been extensively studied for its therapeutic potential in various inflammatory diseases. Research has shown that ANXA1 and its mimetics can effectively limit inflammation in experimental models of myocardial ischemia/reperfusion injury, arthritis, stroke, multiple sclerosis, and sepsis . The protein’s ability to promote the resolution of inflammation makes it a promising target for drug discovery.

Recombinant ANXA1

Recombinant ANXA1 (Mouse) is a form of the protein that is produced through recombinant DNA technology. This involves inserting the gene encoding ANXA1 into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. Recombinant ANXA1 is used in research to study its biological functions and therapeutic potential.

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.