MFGE8 Mouse

Milk Fat Globule-EGF Factor 8 Protein Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

MFGE8 Mouse is implicated in diverse physiological and pathological processes:

Apoptotic Cell Clearance

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 .

Anti-Inflammatory and Organ-Protective Roles

  • 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 .

Dual Role in Acute vs. Chronic Inflammation

  • 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 .

Mechanisms of Action

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 .

Research Tools and Applications

  • 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 .

Clinical and Translational Relevance

  • Biomarker Potential: Serum MFGE8 levels correlate with sepsis severity and heart failure prognosis .

  • Therapeutic Development: rhMFGE8 is being explored for sepsis, wound healing, and neurodegenerative diseases .

Product Specs

Introduction
Milk fat globule-EGF factor 8 protein (Mfge8), also known as lactadherin, is a multifunctional secreted glycoprotein. It plays crucial roles in various biological processes, including: - Mammary gland development (morphogenesis) - Formation of new blood vessels (angiogenesis) - Progression of tumor growth - Maintaining tissue equilibrium (homeostasis) - Suppressing inflammation Mfge8 acts as a link between a molecule found on dying cells (phosphatidylserine) and a receptor on immune cells (Integrin alpha V beta 3). This interaction facilitates the removal of dead and dying cells, contributing to tissue maintenance.
Description
Recombinant MFGE8 Mouse protein, expressed in Sf9 insect cells using a baculovirus system, is a single-chain polypeptide. It undergoes glycosylation, a process of adding sugar molecules, and consists of 413 amino acids (specifically, amino acids 23 to 426). This protein has a molecular weight of 46 kDa, but on SDS-PAGE analysis, it appears as a band around 40-57 kDa. This discrepancy in size is due to glycosylation. For purification and later applications, a 6 amino acid His-tag is added at the C-terminus of the protein. The purification process utilizes proprietary chromatographic techniques to ensure high purity.
Physical Appearance
A clear and colorless solution that has been sterilized by filtration.
Formulation
The provided MFGE8 protein solution has a concentration of 0.25mg/ml. It is formulated in a Phosphate Buffered Saline (PBS) solution with a pH of 7.4 and supplemented with 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep the vial refrigerated at 4°C. For extended storage, freeze the solution at -20°C. To ensure optimal protein stability during long-term storage, consider adding a carrier protein like HSA or BSA at a concentration of 0.1%. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of this MFGE8 protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms

Milk fat globule-EGF factor 8 protein, isoform CRA_a, Putative uncharacterized protein, Mfge8, mCG_6301.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

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.

Q&A

What is MFG-E8 and what are its primary functions in mouse models?

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

  • Supports angiogenesis during wound healing processes

  • 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 .

Where is MFG-E8 expressed in mouse tissues and how does expression change during pathological conditions?

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.

What are the phenotypic characteristics of MFG-E8 knockout mice?

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.

What methods are available for measuring MFG-E8 expression in mouse tissues and biological samples?

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 TypeIntra-Assay PrecisionInter-Assay Precision
Sample12
n2020
Mean (pg/mL)0.5060.936
Standard Deviation0.020.042
CV%44.5

Recovery of MFG-E8 in various matrices shows reliable performance:

Sample TypeAverage % RecoveryRange %
Cell Culture Supernatates (n=4)9990-104
Heparin Plasma (n=4)9490-98
Serum (n=4)10197-104

These methods provide complementary approaches for comprehensive analysis of MFG-E8 expression patterns .

How should researchers design bone marrow transplantation experiments to study MFG-E8 function?

Bone marrow transplantation (BMT) experiments have been instrumental in determining the cell-specific contributions of MFG-E8. When designing such experiments:

  • Donor-recipient combinations:

    • Transplant MFG-E8−/− bone marrow to MFG-E8+/+ mice to study the impact of macrophage-derived MFG-E8 deficiency

    • Transplant wild-type bone marrow to MFG-E8−/− mice to assess whether normal macrophages can rescue the phenotype

  • 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 .

What approaches can be used to study MFG-E8's role in mediating cell-cell interactions?

Several methodological approaches have proven effective for investigating MFG-E8-mediated cellular interactions:

  • Receptor inhibition strategies:

    • MFGE8 RGD domain ablation to disrupt integrin-binding capability

    • Competitive RGD-peptide inhibition to block integrin binding

    • Antibody masking of alpha V integrin receptors to prevent MFG-E8 binding

  • 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 .

How does MFG-E8 deficiency affect wound healing processes in mouse models?

MFG-E8 deficiency significantly impairs multiple aspects of wound healing in mouse models:

  • Efferocytosis and inflammation:

    • MFG-E8−/− mice exhibit impaired clearance of apoptotic cells at wound sites

    • This impairment leads to exaggerated and persistent inflammatory responses

    • Prolonged inflammation delays progression to the proliferative phase of wound healing

  • Angiogenesis impairment:

    • Wound macrophage-derived MFG-E8 serves as a critical driver of wound angiogenesis

    • MFG-E8−/− mice display poor wound vascularization

    • Transplantation studies confirm the importance of macrophage-derived MFG-E8 for proper wound vascularization

  • Diabetic wound complications:

    • Hyperglycemia and advanced glycation end products inactivate MFG-E8

    • Diabetic db/db mice show impaired efferocytosis with persistent inflammation

    • This mechanism provides insight into the poor wound healing characteristic of diabetes

  • Rescue potential:

    • Wild-type bone marrow transplantation to MFG-E8−/− mice improves wound closure and vascularization

    • This suggests potential therapeutic approaches for wound healing disorders

These findings establish MFG-E8 as a multifunctional mediator in wound healing that links efferocytosis, inflammation resolution, and angiogenic processes.

What is the role of MFG-E8 in inflammatory conditions such as acute pancreatitis?

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:

    • Knocking out Mfge8 in mice exacerbates the severity of cerulein-induced acute pancreatitis

    • MFG-E8−/− mice show delayed resolution of pancreatic inflammation

    • The inflammatory injury is more pronounced and persists longer compared to wild-type mice

  • Therapeutic potential:

    • Administration of recombinant MFG-E8 attenuates cerulein-induced acute pancreatitis

    • Recombinant MFG-E8 promotes repair of pancreatic injury in Mfge8 knockout mice

    • These findings suggest MFG-E8 as a novel therapeutic target for acute pancreatitis

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.

How does MFG-E8 influence cardiac pathologies in mouse models?

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:

    • MFGE8 alleviates pressure overload-induced cardiac hypertrophy in mice

    • This protective effect occurs through inhibition of the Akt (protein kinase B)-dependent pathway

    • MFGE8 regulates the Akt–GSK-3β/mTOR signaling pathway in cardiomyocytes

  • Cross-species validation:

    • An Mfge8-knockout rat line has been generated to validate findings

    • The protective effect of MFGE8 against pathological cardiac hypertrophy is conserved across species

  • Therapeutic implications:

    • Administration of recombinant human MFGE8 (rhMFGE8) significantly reverses aortic banding-triggered cardiac hypertrophy in mice

    • MFGE8 may serve as both a biomarker for heart failure diagnosis and a therapeutic target

These findings establish MFGE8 as an endogenous negative regulator of pathological cardiac hypertrophy with potential diagnostic and therapeutic applications.

What mechanisms explain the development of autoimmunity in MFG-E8-deficient mice?

The development of autoimmunity in MFG-E8-deficient mice involves multiple interconnected mechanisms:

  • Defective apoptotic cell clearance:

    • MFG-E8 functions as an opsonin that facilitates phagocytosis of apoptotic cells

    • MFG-E8 deficiency leads to delayed clearance of dying cells, resulting in secondary necrosis and release of immunogenic intracellular contents

  • 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:

    • MFG-E8−/− mice show striking activation of effector memory CD8+ T cells

    • CD8+ T cell responses to both exogenous and endogenous apoptotic cell-associated antigens are enhanced

    • Older MFG-E8−/− mice spontaneously develop dermatitis associated with CD8+ T cell infiltration

  • 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.

How can researchers distinguish between direct and indirect effects of MFG-E8 deficiency in experimental models?

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.

What approaches are most effective for investigating the therapeutic potential of recombinant MFG-E8?

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:

    • Complement in vivo studies with ex vivo and in vitro approaches

    • Verify engagement of expected signaling pathways (e.g., Akt inhibition in cardiac models)

    • Confirm cellular targets through tissue-specific marker analysis

These methodological considerations provide a framework for rigorous evaluation of recombinant MFG-E8 as a therapeutic agent across multiple disease models.

What are the critical considerations for studying MFG-E8-mediated extracellular vesicle interactions?

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:

    • Develop traceable cell culture models as demonstrated in sperm-EV interaction studies

    • Consider 3D co-culture systems to better recapitulate in vivo environments

    • Validate findings in appropriate animal models

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.

How should researchers interpret contradictory findings regarding MFG-E8 function across different disease models?

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.

Product Science Overview

Introduction

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.

Structure and Function

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 .

Biological Roles
  1. Phagocytosis and Apoptosis: MFG-E8 facilitates the recognition and engulfment of apoptotic cells by macrophages, promoting the clearance of dying cells and preventing the release of inflammatory substances .
  2. Angiogenesis: MFG-E8 promotes the formation of new blood vessels, which is essential for tissue repair and regeneration .
  3. Immune Regulation: MFG-E8 has been shown to modulate immune responses, reducing inflammation and improving outcomes in various disease models .
Therapeutic Potential

The recombinant form of MFG-E8 has shown promise in several therapeutic applications:

  • Inflammatory Diseases: By enhancing the clearance of apoptotic cells, MFG-E8 can reduce inflammation and improve prognosis in conditions such as sepsis and acute lung injury .
  • Cardiovascular Diseases: MFG-E8 has been identified as a potential biomarker and therapeutic target for heart failure and cardiac hypertrophy .
  • Cancer: MFG-E8’s role in angiogenesis makes it a potential target for cancer therapy, as inhibiting its function could reduce tumor growth and metastasis .

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.