MFGE8 Mouse is implicated in diverse physiological and pathological processes:
MFGE8 acts as a "bridging molecule" between PS on apoptotic cells and phagocyte integrins, facilitating their engulfment. Deficiency in MFGE8 leads to autoimmune disorders due to accumulation of apoptotic debris . For example:
Autoimmunity: MFGE8-knockout mice develop splenomegaly, glomerulonephritis, and anti-dsDNA antibodies .
Neuroprotection: Recombinant MFGE8 reduces neuronal apoptosis in traumatic brain injury models by activating PI3K-Akt signaling .
Sepsis: MFGE8 treatment suppresses hepatic oxidative stress and ferroptosis in septic mice, improving 28-day survival rates .
Pancreatitis: MFGE8 deficiency exacerbates cerulein-induced pancreatic injury, while recombinant MFGE8 accelerates acinar repair .
Cardiac Hypertrophy: MFGE8 inhibits Akt-GSK-3β/mTOR pathways, attenuating pathological cardiac remodeling .
Acute Phase: MFGE8 suppresses pro-inflammatory cytokines (e.g., TNF-α, IL-6) and oxidative stress .
Chronic Phase: Prolonged MFGE8 elevation exacerbates neuronal loss in ischemic brain injury by promoting phagocytosis of viable neurons .
MFGE8 Mouse modulates signaling pathways critical for inflammation and tissue repair:
Akt Pathway: MFGE8 inhibits Akt activation in cardiac hypertrophy but activates it in neuroprotection .
Nrf2 Pathway: Enhances antioxidant responses by upregulating glutathione (GSH) and superoxide dismutase (SOD) .
Integrin Signaling: Promotes VEGF-dependent angiogenesis and macrophage polarization toward anti-inflammatory (M2) phenotypes .
ELISA Kits: Quantify MFGE8 levels in serum or cell culture supernatants (e.g., R&D Systems Mouse MFGE8 Quantikine ELISA Kit) .
Antibodies: Used for Western blot, IHC, and flow cytometry (e.g., Clone 340614 targeting Ala23-Cys463) .
Therapeutic Testing: Recombinant MFGE8 is administered intraperitoneally or intracerebroventricularly in murine models .
Milk fat globule-EGF factor 8 protein, isoform CRA_a, Putative uncharacterized protein, Mfge8, mCG_6301.
ADLASGDFCD SSLCLNGGTC LTGQDNDIYC LCPEGFTGLV CNETERGPCS PNPCYNDAKC LVTLDTQRGD IFTEYICQCP VGYSGIHCET GCSTQLGMEG GAIADSQISA SSVYMGFMGL QRWGPELARL YRTGIVNAWT ASNYDSKPWI QVNLLRKMRV SGVMTQGASR AGRAEYLKTF KVAYSLDGRK FEFIQDESGG DKEFLGNLDN NSLKVNMFNP TLEAQYIKLY PVSCHRGCTL RFELLGCELH GCSEPLGLKN NTIPDSQMSA SSSYKTWNLR AFGWYPHLGR LDNQGKINAW TAQSNSAKEW LQVDLGTQRQ VTGIITQGAR DFGHIQYVAS YKVAHSDDGV QWTVYEEQGS SKVFQGNLDN NSHKKNIFEK PFMARYVRVL PVSWHNRITL RLELLGCHHH HHH.
MFG-E8 is a secreted glycoprotein originally identified in lactating mammary glands but now recognized to be expressed in multiple tissues. In mouse models, MFG-E8 serves several critical functions:
Functions as an opsonin by binding phosphatidylserine on apoptotic cells, facilitating their phagocytic clearance (efferocytosis)
Plays a vital role in resolving inflammation and promoting tissue repair
Provides protection against inflammatory tissue injury in multiple organ systems
Mediates intercellular communication, including the interaction between spermatozoa and extracellular vesicles
Research using MFG-E8-knockout mice has demonstrated that absence of this protein leads to impaired efferocytosis, exaggerated inflammatory responses, and development of autoimmune conditions resembling lupus .
MFG-E8 exhibits both constitutive and inducible expression patterns in mouse tissues:
Pancreas: Constitutively expressed and increases during cerulein-induced acute pancreatitis
Pancreatic cell types: In situ hybridization has revealed that ductal epithelial cells express Mfge8 transcripts at baseline, while during pancreatitis, expression extends to acinar cells and endothelial cells
Immune cells: Activated macrophages and immature dendritic cells secrete MFG-E8
Male reproductive tract: Expressed in the epididymis where it mediates sperm-extracellular vesicle interactions
Cardiovascular system: Circulating levels decrease significantly in cardiac pathological conditions, with progressive decline correlating with worsening heart function
The dynamic regulation of MFG-E8 expression during pathological states suggests its role as a responsive mediator during tissue injury and repair processes.
MFG-E8 knockout mice (MFG-E8−/−) display several distinctive phenotypic characteristics:
Wound healing: Impaired efferocytosis associated with exaggerated inflammatory responses, poor angiogenesis, and delayed wound closure
Autoimmunity: Development of lupus-like autoimmunity with production of class-switched IgG autoantibodies
Immune dysregulation: Older MFG-E8−/− mice show striking activation of effector memory CD8+ T cells and develop spontaneous dermatitis with CD8+ T cell infiltration
Pancreatic inflammation: Exacerbated severity and delayed resolution of cerulein-induced acute pancreatitis
Altered T cell populations: Significant downregulation of CD62L in both CD4+ and CD8+ T cell populations by 4 months of age, with persistent alterations in the ratio of effector/central memory CD8+ T cells throughout aging
Enhanced cytokine production: Upon activation, CD8+ T cells from older MFG-E8−/− mice produce significantly more IFN-γ and IL-2 than wild-type mice
These phenotypes demonstrate the multifaceted role of MFG-E8 in maintaining tissue homeostasis and proper immune function.
Researchers have several methodological options for quantifying MFG-E8 in mouse samples:
ELISA: The Quantikine Mouse MFG-E8 Immunoassay provides a solid-phase ELISA designed to measure mouse MFG-E8 in cell culture supernatants, serum, and plasma with high precision
Immunoblot analysis: Western blotting has been effectively used to demonstrate constitutive expression of MFG-E8 in mouse pancreas and its upregulation during pancreatitis
In situ hybridization: This technique allows visualization of Mfge8 transcripts in specific cell types within tissues, as demonstrated in pancreatic studies
Immunohistochemistry: Used to localize MFG-E8 protein in tissue sections
Performance characteristics of the Quantikine Mouse MFG-E8 ELISA demonstrate high precision:
Test Type | Intra-Assay Precision | Inter-Assay Precision |
---|---|---|
Sample | 1 | 2 |
n | 20 | 20 |
Mean (pg/mL) | 0.506 | 0.936 |
Standard Deviation | 0.02 | 0.042 |
CV% | 4 | 4.5 |
Recovery of MFG-E8 in various matrices shows reliable performance:
Sample Type | Average % Recovery | Range % |
---|---|---|
Cell Culture Supernatates (n=4) | 99 | 90-104 |
Heparin Plasma (n=4) | 94 | 90-98 |
Serum (n=4) | 101 | 97-104 |
These methods provide complementary approaches for comprehensive analysis of MFG-E8 expression patterns .
Bone marrow transplantation (BMT) experiments have been instrumental in determining the cell-specific contributions of MFG-E8. When designing such experiments:
Donor-recipient combinations:
Technical considerations:
Irradiate recipient mice (typically with 9-10 Gy) prior to BMT
Inject bone marrow cells (typically 5-10×10^6 cells) via tail vein
Allow 6-8 weeks for reconstitution before experimental interventions
Verify chimerism through flow cytometry of peripheral blood cells
Experimental readouts:
For wound healing studies, employ splinted full-thickness excisional wound models
Measure wound closure rates, vascular density, inflammatory marker expression, and efferocytosis efficiency
Analyze tissue-resident versus bone marrow-derived macrophage populations
Research has demonstrated that transplantation of MFG-E8−/− bone marrow to wild-type mice results in impaired wound closure and compromised wound vascularization, while MFG-E8−/− mice receiving wild-type bone marrow showed improved wound closure and vascularization .
Several methodological approaches have proven effective for investigating MFG-E8-mediated cellular interactions:
Receptor inhibition strategies:
Tracking extracellular vesicle interactions:
Labeling extracellular vesicles with fluorescent markers to visualize uptake
Co-culture systems with differentially labeled cell populations
Live-cell imaging to track vesicle trafficking
Functional assays:
Assess macromolecular cargo delivery by measuring the redistribution of labeled proteins
Quantify phagocytosis efficiency of apoptotic cells in the presence or absence of MFG-E8
Analyze intracellular processing pathways using endosomal/lysosomal markers
These complementary approaches have successfully demonstrated MFG-E8's critical role in mediating interactions between mouse sperm and extracellular vesicles, with the RGD tripeptide motif being particularly important for binding to integrin receptors .
MFG-E8 deficiency significantly impairs multiple aspects of wound healing in mouse models:
Efferocytosis and inflammation:
Angiogenesis impairment:
Diabetic wound complications:
Rescue potential:
These findings establish MFG-E8 as a multifunctional mediator in wound healing that links efferocytosis, inflammation resolution, and angiogenic processes.
MFG-E8 plays a protective role in acute pancreatitis through several mechanisms:
Expression patterns during pancreatitis:
MFG-E8 is constitutively expressed in the murine pancreas
Its expression increases during cerulein-induced acute pancreatitis
In situ hybridization reveals that ductal epithelial cells express Mfge8 transcripts at baseline
During pancreatitis, expression expands to include acinar cells and endothelial cells
Impact of MFG-E8 deficiency:
Therapeutic potential:
The research conclusively demonstrates that MFG-E8 protects the pancreas against inflammatory injury and promotes pancreatic tissue repair, representing a potential therapeutic avenue for managing acute pancreatitis.
MFG-E8 demonstrates important regulatory functions in cardiac pathology:
Clinical correlations:
Circulating MFGE8 levels decrease remarkably in patients with dilated cardiomyopathy (DCM)
Serum MFGE8 levels decline progressively with worsening heart failure (New York Heart Association classification)
Levels negatively correlate with the severity of cardiac functional impairment and degree of cardiac remodeling
Mechanistic insights:
Cross-species validation:
Therapeutic implications:
These findings establish MFGE8 as an endogenous negative regulator of pathological cardiac hypertrophy with potential diagnostic and therapeutic applications.
The development of autoimmunity in MFG-E8-deficient mice involves multiple interconnected mechanisms:
Defective apoptotic cell clearance:
Altered antigen processing and presentation:
MFG-E8 controls both phagocytic ingestion of cell fragments and their intracellular processing into MHC-antigen complexes
In MFG-E8−/− mice, smaller apoptotic cell fragments persist in dendritic cell endosomal compartments for extended periods (24 hours)
This leads to enhanced cross-presentation of self-antigens to CD8+ T cells
Enhanced CD8+ T cell responses:
T cell subset imbalance:
Increased ratios of effector/central memory CD44+CD8+ T cells are observed in MFG-E8−/− mice
CD8+ T cells from MFG-E8−/− mice produce significantly more IFN-γ and IL-2 upon activation
The CD8+ population shows more striking activation profiles, along with relative depletion in older mice, likely due to activation-induced cell death
These mechanisms demonstrate that MFG-E8 deficiency promotes immune responses to self-antigens through both impaired clearance of dying cells and altered intracellular processing, leading to enhanced self-antigen presentation.
Distinguishing direct from indirect effects of MFG-E8 deficiency requires sophisticated experimental approaches:
Tissue-specific knockout models:
Generate conditional MFG-E8 knockout mice using Cre-loxP technology
Target specific cell types (e.g., macrophages, dendritic cells, epithelial cells) to determine cell-specific contributions
Compare phenotypes between global and conditional knockouts to identify secondary effects
Chimeric approaches:
Bone marrow transplantation experiments between wild-type and MFG-E8−/− mice
This approach has successfully demonstrated that macrophage-derived MFG-E8 is critical for wound angiogenesis
When MFG-E8−/− bone marrow was transplanted to wild-type mice, impaired wound closure and compromised wound vascularization resulted
Conversely, wild-type bone marrow transplanted to MFG-E8−/− mice improved these parameters
Temporal control systems:
Employ inducible knockout systems (e.g., tamoxifen-inducible Cre) to eliminate developmental confounders
This allows examination of acute versus chronic adaptations to MFG-E8 deficiency
Complementation studies:
Administer recombinant MFG-E8 protein to knockout models
Determine which phenotypes can be rescued and which cannot
Varying dosage and timing of administration can reveal threshold-dependent effects
Studies have shown that recombinant MFG-E8 administration attenuates cerulein-induced acute pancreatitis and promotes repair in knockout mice
These approaches collectively enable researchers to parse the complex network of direct and indirect effects resulting from MFG-E8 deficiency.
Investigation of recombinant MFG-E8 as a therapeutic agent requires systematic methodological approaches:
Production and characterization:
Expression systems (bacterial, mammalian, or insect cells) must be optimized for proper post-translational modifications
Purification protocols should ensure elimination of endotoxin contamination
Biological activity verification through binding assays and functional tests is essential
Dosage optimization studies:
Conduct dose-response experiments to determine minimum effective dose
Establish pharmacokinetic profiles including half-life and tissue distribution
Determine optimal administration routes (intravenous, intraperitoneal, local application)
Timing considerations:
Test preventive versus therapeutic administration protocols
In acute pancreatitis models, administration of recombinant MFG-E8 has successfully attenuated cerulein-induced inflammation
For cardiac hypertrophy, recombinant human MFG-E8 significantly reversed aortic banding-triggered cardiac hypertrophy
Disease-specific considerations:
For wound healing: assess closure rates, vascularization, inflammatory cell infiltration
For autoimmune conditions: monitor autoantibody production, T cell activation markers
For acute inflammatory conditions: measure tissue damage biomarkers, inflammatory cytokines
For cardiac pathologies: evaluate cardiac function, hypertrophy markers, and remodeling
Mechanistic validation:
These methodological considerations provide a framework for rigorous evaluation of recombinant MFG-E8 as a therapeutic agent across multiple disease models.
Investigating MFG-E8's role in extracellular vesicle (EV) interactions requires specialized approaches:
EV isolation and characterization:
Employ differential ultracentrifugation, size exclusion chromatography, or commercial isolation kits
Verify EV size distribution using nanoparticle tracking analysis or dynamic light scattering
Confirm EV marker proteins (CD9, CD63, CD81) through Western blotting
Ensure consistent EV preparations across experiments
Binding domain analysis:
The RGD (Arg-Gly-Asp) tripeptide motif in MFG-E8 is crucial for binding to integrin receptors
Research has shown that MFGE8 RGD domain ablation significantly inhibits the uptake of EV-delivered proteins
Complementary approaches include competitive RGD-peptide inhibition and antibody-masking of alpha V integrin receptors
Tracking methodologies:
Label EVs with lipophilic dyes or fluorescent protein tags
Employ live-cell imaging to visualize EV-cell interactions in real-time
Use confocal microscopy to determine intracellular localization of EV cargo
Functional readouts:
Assess the efficiency of macromolecular cargo transfer
Evaluate functional consequences of delivered proteins or RNAs
Measure physiological responses in recipient cells (e.g., signaling pathway activation)
Model systems:
These methodological considerations provide a framework for investigating the complex role of MFG-E8 in mediating EV interactions, which underlie various physiological and pathological processes.
Interpreting contradictory findings about MFG-E8 function requires careful consideration of several factors:
By considering these factors and employing rigorous experimental designs, researchers can better interpret and reconcile apparently contradictory findings regarding MFG-E8 function across different disease models and experimental systems.
Milk Fat Globule-EGF Factor 8 (MFG-E8), also known as lactadherin, is a glycoprotein that plays a crucial role in various cellular processes, including cell signaling, apoptosis, and phagocytosis . The recombinant form of MFG-E8 derived from mouse models is particularly valuable for researchers studying its biological functions and therapeutic potential.
MFG-E8 is characterized by its unique structure, which includes two epidermal growth factor (EGF)-like domains and a discoidin-like domain. The EGF-like domains are responsible for binding to integrins, while the discoidin-like domain binds to phosphatidylserine on apoptotic cells . This dual binding capability allows MFG-E8 to mediate the clearance of apoptotic cells by phagocytes, thereby maintaining tissue homeostasis and preventing inflammation .
The recombinant form of MFG-E8 has shown promise in several therapeutic applications: