IL12 Human, His

Interleukin 12 His Tag Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IL-12 Human, His

IL-12 Human, His refers to recombinant human interleukin-12 (IL-12) engineered with a histidine (His) tag for efficient purification via immobilized metal affinity chromatography (IMAC). This cytokine is a heterodimer composed of two subunits: p35 (IL-12A) and p40 (IL-12B), which form the bioactive p70 complex. IL-12 is critical for initiating Th1 immune responses, enhancing cytotoxic T and NK cell activity, and modulating anti-tumor and anti-infectious immunity .

Immune Modulation

IL-12 drives the differentiation of naïve T cells into Th1 cells, characterized by IFN-γ production. Key activities include:

  • IFN-γ Induction: Synergizes with IL-2 and IL-18 to upregulate IFN-γ in NK and T cells .

  • Cytotoxicity Enhancement: Increases perforin/granzyme expression in CD8+ T cells and NK cells .

  • Anti-Angiogenic Effects: Induces IP-10 (CXCL10) to block angiogenesis .

Receptor Signaling

Receptor SubunitsFunctionSource
IL-12Rβ1Binds p40; part of IL-23 and IL-35 receptors
IL-12Rβ2Binds p35; critical for STAT4 phosphorylation
STAT4 ActivationDrives Th1 differentiation and IFN-γ/TNF-α production

IL-12Rβ2 expression is upregulated by antigen presentation and IFN-γ, while IL-4 suppresses it .

Preclinical Studies

ModelOutcomeSource
Tumor ModelsNHS-IL12 (tumor-targeted IL-12) reduces MDSCs/TAMs, enhances CD8+ T-cell infiltration
NeuroinflammationIL-12R signaling in neurons reduces CNS damage during EAE (multiple sclerosis model)
Viral InfectionsSynergizes with IL-18 to boost IFN-γ in macrophages and B cells

Clinical Trials

Phase/TargetOutcomeSource
Phase I (Solid Tumors)NHS-IL12 (16.8 µg/kg) induced IFN-γ/TNF-α; MTD achieved with transient toxicity
Combination TherapyNHS-IL12 + avelumab showed clinical responses in bladder cancer
Hepatic ToxicityElevated ALT/AST observed in early trials; mitigated via targeted delivery

IL-12 Family Cytokines: Comparative Analysis

CytokineSubunitsPrimary FunctionSource
IL-12p35 + p40Th1 differentiation, anti-tumor
IL-23p19 + p40Th17 differentiation, autoimmunity
IL-27p28 + EBI3Treg activation, immunosuppression
IL-35p35 + EBI3Anti-inflammatory, tolerance induction

Therapeutic Challenges and Innovations

  • Toxicity: Systemic IL-12 causes cytokine release syndrome (CRS); strategies include tumor-targeted delivery (e.g., NHS-IL12) and recombinant protein engineering .

  • Neuroprotection: IL-12Rβ2 expression in neurons mitigates neurodegeneration by upregulating trophic factors .

  • Biomarker Potential: Baseline monocyte/plasmacytoid DC levels correlate with clinical response in trials .

Product Specs

Introduction
Interleukin-12 (IL-12) is a heterodimeric cytokine that plays a crucial role in the immune system by stimulating the production of interferon gamma (IFN-γ) from T-cells and natural killer (NK) cells. Additionally, IL-12 drives the differentiation of T helper 1 (Th1) cells, establishing its importance in initiating cell-mediated immunity.
Description
Recombinant human IL-12, expressed in a baculovirus system, is a glycosylated heterodimer linked by disulfide bonds. The heterodimer comprises two subunits: IL12A and IL12B. IL12A, with a molecular weight of 23.3 kDa, consists of amino acids 23-219 (totaling 203 amino acids). IL12B, with a molecular weight of 34.6 kDa, is composed of amino acids 23-328 (totaling 306 amino acids). The predicted molecular weight of the complete IL-12 heterodimer is 57.9 kDa. However, the apparent molecular weight on SDS-PAGE may appear higher. For purification, a 6-amino acid His-Tag is fused to the C-terminus of IL12A, and proprietary chromatographic techniques are employed.
Physical Appearance
A clear and colorless solution, devoid of any particulate matter.
Formulation
The IL-12 protein is supplied in a solution containing Phosphate buffered saline (PBS) at a pH of 7.4 and 10% glycerol. The protein concentration is 0.5 mg/ml.
Stability
For short-term storage (2-4 weeks), the IL-12 solution can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is advised for long-term storage. To maintain protein integrity, avoid repeated freeze-thaw cycles.
Purity
The purity of the IL-12 protein is greater than 90% as determined by SDS-PAGE analysis.
Biological Activity
The biological activity of IL-12 is evaluated by measuring its ability to induce IFN-γ production in NK-92 cells using an IFN-γ ELISA. The effective dose that results in 50% activity (ED50) is less than or equal to 0.03 ng/ml.
Synonyms
23-219aa (IL12A) / 23-328aa (IL12B), Interleukin 12 (subunit beta/alpha), IL12 His, NKSF1, NKSF, CTL maturation factor (TCMF), Cytotoxic lymphocyte maturation factor (CLMF), TSF, Edodekin-alpha, IL-12.
Source
Baculovirus.
Amino Acid Sequence
IL12A(p35):
RNLPVATPDP GMFPCLHHSQ NLLRAVSNML QKARQTLEFY PCTSEEIDHE DITKDKTSTV EACLPLELTK NESCLNSRET SFITNGSCLA SRKTSFMMAL CLSSIYEDLK MYQVEFKTMN AKLLMDPKRQ IFLDQNMLAV IDELMQALNF NSETVPQKSS LEEPDFYKTK IKLCILLHAF RIRAVTIDRV MSYLNASHHH HHH.

IL12B(p40):
IWELKKDVYV VELDWYPDAP GEMVVLTCDT PEEDGITWTL DQSSEVLGSG KTLTIQVKEF GDAGQYTCHK GGEVLSHSLL LLHKKEDGIW STDILKDQKE PKNKTFLRCE AKNYSGRFTC WWLTTISTDL TFSVKSSRGS SDPQGVTCGA ATLSAERVRG DNKEYEYSVE CQEDSACPAA EESLPIEVMV DAVHKLKYEN YTSSFFIRDI IKPDPPKNLQ LKPLKNSRQV EVSWEYPDTW STPHSYFSLT FCVQVQGKSK REKKDRVFTD KTSATVICRK NASISVRAQD RYYSSSWSEW ASVPCS.

Q&A

What is the molecular structure of human IL-12 and how does His-tagging affect its function?

Human IL-12 is a heterodimeric cytokine composed of two glycosylated and disulfide-linked subunits: a lighter p35 subunit (IL-12A, approximately 34 kDa) covalently linked to a heavier p40 subunit (IL-12B, approximately 42 kDa) . Together they form a functional 57 kDa heterodimer (when non-reduced) . The p35 subunit is encoded by gene ID 3592, while p40 is encoded by gene ID 3593 .

  • Binding assays to IL-12 receptor components (IL-12Rβ1 and IL-12Rβ2)

  • IFNγ induction assays using NK cells or T cells

  • STAT4 phosphorylation assays in responsive cells

The specific activity of properly folded His-tagged human IL-12 should be approximately 3.00 × 10^6 IU/mg with activity detectable at concentrations of 0.8-4 ng/mL .

How does recombinant human IL-12 differ from native IL-12 in terms of glycosylation and folding?

Recombinant human IL-12 production quality depends greatly on the expression system used:

Expression SystemGlycosylation PropertiesAdvantagesConsiderations
HEK293 cellsAuthentic human-like glycosylation patternMost similar to native human IL-12, proper foldingHigher production cost, lower yield
CHO cellsMammalian glycosylation with minor differencesGood bioactivity, scalableSome glycoform differences
E. coliNo glycosylationCost-effective, high yieldRefolding often required, potential immunogenicity
Insect cellsSimpler glycosylation patternHigh yield, some post-translational modificationsDifferent glycan structures than human

High-quality recombinant human IL-12 should demonstrate >95% purity with endotoxin levels <1 EU/μg . The alpha chain (p35) shows more acidic pI patterns in the secreted heterodimer compared to intracellular forms, indicating post-translational modifications occur during secretion . When evaluating recombinant IL-12 preparations, researchers should verify both correct heterodimer formation and proper glycosylation, as these significantly impact biological activity.

What are the optimal reconstitution and storage conditions for His-tagged human IL-12?

For optimal handling of lyophilized His-tagged human IL-12:

Reconstitution protocol:

  • Briefly centrifuge the vial before opening to collect all material

  • Reconstitute to 0.2-0.25 mg/mL in sterile 1× PBS (pH 7.4)

  • For increased stability, add 0.1% endotoxin-free recombinant human serum albumin (HSA)

  • Gently swirl or tap the vial to mix; avoid vortexing which can damage protein structure

  • Allow complete reconstitution (approximately 10-15 minutes) before aliquoting

Storage conditions:

  • Lyophilized protein: Stable for 1 year when stored at -20°C to -80°C

  • Reconstituted protein:

    • Short-term (≤1 week): 4°C

    • Medium-term (≤1 month): -20°C in single-use aliquots

    • Long-term (≤6 months): -80°C in single-use aliquots

Critical considerations:

  • Repeated freeze-thaw cycles dramatically reduce activity; create single-use aliquots

  • Some preparations benefit from carrier proteins like 0.1% HSA or 0.1% BSA to prevent adsorption to plastic surfaces

  • Verify activity after extended storage with functional assays

  • Record lot-specific activity to account for batch-to-batch variations

How can researchers accurately measure the biological activity of His-tagged human IL-12?

Multiple complementary assays should be employed to assess the biological activity of His-tagged human IL-12:

1. Receptor binding assays:

  • ELISA-based binding assays using recombinant IL-12Rβ1 (ED₅₀ typically between 3-30 ng/mL)

  • Surface plasmon resonance (SPR) to determine binding kinetics to both receptor subunits

2. Cell-based functional assays:

  • NK cell activation: Measure IFNγ production by human NK cells (PBMC-derived or NK cell lines)

  • T cell proliferation: Assess proliferation of IL-12-responsive T cell subsets

  • STAT4 phosphorylation: Western blot or flow cytometry detection of phospho-STAT4 in responsive cells

3. Cytokine induction cascade:

  • Quantify secondary cytokine production (IFNγ, TNFα, IP-10) by ELISA or multiplex assays

  • Monitor upregulation of activation markers on immune cells by flow cytometry

Standardization approaches:

  • Include a reference standard with known activity in each assay

  • Express activity in International Units (IU) where 1 IU equals the amount inducing half-maximal biological response

  • A high-quality preparation should show specific activity of approximately 3.00 × 10^6 IU/mg

When comparing different His-tagged IL-12 preparations, researchers should use the same assay system to avoid methodological variations that could affect activity measurements.

How can researchers design IL-12 variants with attenuated activity for reduced toxicity while maintaining therapeutic efficacy?

Designing attenuated IL-12 variants requires strategic modifications to receptor binding interfaces while preserving essential biological functions:

Computational modeling approaches:

  • Generate high-resolution computational models of the human IL-12:receptor complex

  • Identify key amino acid residues at receptor-binding interfaces through molecular dynamics simulations

  • Prioritize mutations that modulate receptor affinity without disrupting protein folding

Strategic modification targets:

  • Mutations at the IL-12p35:IL-12Rβ2 interface can selectively reduce NK cell activation while preserving T cell responses

  • Modifications to the IL-12p40:IL-12Rβ1 interface generally affect all IL-12 signaling

  • Engineering glycosylation sites at strategic positions can sterically hinder receptor binding

Validation experiments:

  • Differential binding assays to IL-12Rβ1 versus IL-12Rβ2

  • Comparative cell-based assays using purified NK cells versus T cells

  • In vivo models comparing anti-tumor efficacy versus systemic cytokine release

Recent studies have successfully created IL-12 variants with reduced NK cell activation that maintain T cell responses. This approach addresses the challenge of systemic toxicity while preserving anti-tumor efficacy, as NK cell-derived IFNγ is implicated in dose-limiting toxicities in clinical settings .

What experimental approaches can address the pharmacokinetic limitations of recombinant human IL-12 therapy?

Several innovative approaches can improve the pharmacokinetic profile of recombinant human IL-12:

Delivery route optimization:

  • Intravenous administration has shown superior response rates (40% partial/complete response) compared to subcutaneous delivery (7% response) at equivalent toxicity profiles in lymphoma patients

  • Intratumoral delivery concentrates activity at target sites while reducing systemic exposure

  • Intraperitoneal administration for localized treatment of ovarian or peritoneal cancers

Structural modifications:

  • PEGylation to increase half-life and reduce immunogenicity

  • Fc-fusion proteins for extended circulation time

  • Site-specific modifications that preserve receptor binding while improving stability

Advanced delivery systems:

  • Encapsulation in liposomes or nanoparticles for controlled release

  • Tumor-targeted antibody-cytokine fusion proteins

  • Cell-based delivery systems using engineered immune cells

Combinatorial approaches:

  • Co-administration with agents that mitigate IL-12-induced toxicity

  • Sequential administration protocols to build tolerance to IL-12

  • Dose-escalation strategies with careful monitoring of cytokine responses

Each approach requires careful validation of both pharmacokinetic parameters and biological activity, as modifications can affect IL-12's heterodimeric structure and receptor binding properties .

How can researchers accurately distinguish between active heterodimeric IL-12 and inactive individual subunits in recombinant preparations?

Distinguishing active IL-12 heterodimers from individual subunits requires multiple analytical approaches:

Protein-level analysis:

  • Non-reducing vs. reducing SDS-PAGE: Under non-reducing conditions, intact IL-12 heterodimer appears as a ~57 kDa band, while reducing conditions reveal separate p35 (~34 kDa) and p40 (~42 kDa) subunits

  • Size exclusion chromatography (SEC): Separates heterodimer from free p40 subunit and p40 homodimers

  • Western blotting: Using antibodies specific to conformational epitopes present only in the heterodimer

Functional verification:

  • Receptor binding assays using both IL-12Rβ1 and IL-12Rβ2 components

  • Cell-based assays measuring STAT4 phosphorylation (only functional heterodimer activates this pathway)

  • IFNγ induction in NK or T cells as a biomarker of active IL-12

Critical considerations:

  • Free p40 is typically produced in excess and can form homodimers with antagonistic activity

  • The p35 subunit is poorly secreted alone and requires co-expression with p40

  • Acidic pI patterns differ between secreted and intracellular heterodimers, indicating post-translational modifications during secretion

For researchers developing or using His-tagged IL-12, validation of heterodimer integrity is essential, as expression systems may produce varying amounts of free p40 that can interfere with experimental results.

What are the key differences in experimental outcomes when using different expression systems for producing His-tagged human IL-12?

Expression system selection significantly impacts His-tagged human IL-12 quality and experimental outcomes:

Expression SystemImpact on QualityExperimental ConsiderationsBest Applications
HEK293 cells- Authentic glycosylation
- Proper disulfide formation
- Endotoxin-free
- Animal component-free possible
- Higher cost
- More physiologically relevant outcomes
- >95% active heterodimer
- In vivo studies
- Clinical research
- Sensitive immunological assays
CHO cells- Good mammalian glycosylation
- Proper folding
- Scalable production
- Different glycoform profile than human
- Potential for clone-dependent variability
- Large-scale production
- Most in vitro applications
E. coli- No glycosylation
- Often requires refolding
- Higher proportion of inactive protein
- Batch-to-batch variability
- Potential endotoxin contamination
- Lower specific activity
- Structural studies
- Applications where glycosylation is not critical
Baculovirus/insect cells- Partial glycosylation
- Good folding
- High yield
- Non-mammalian glycosylation patterns
- Different bioactivity profile
- High-yield requirements
- Applications where exact glycosylation is less critical

Critical research implications:

  • Glycosylation affects IL-12 stability, half-life, and immunogenicity

  • The acidic profile of secreted p35 differs from intracellular p35, suggesting essential post-translational modifications during secretion

  • IL-12 from HEK293 cells shows activity at 0.8-4 ng/mL with specific activity of >3 × 10^6 IU/mg

  • Endotoxin contamination (<1 EU/μg standard) is critical as it can confound immunological experiments

Researchers should select expression systems based on their specific experimental requirements, with HEK293-expressed IL-12 representing the gold standard for immunological and in vivo studies due to its authentic human glycosylation patterns .

How are engineered IL-12 variants being designed to overcome clinical limitations in cancer immunotherapy?

Recent advances in engineered IL-12 variants address key clinical limitations through several innovative strategies:

Receptor binding modifications:

  • Computational modeling of the IL-12:receptor complex enables design of variants with attenuated NK cell activation but preserved T cell responses

  • Strategic mutations that reduce IFNγ-associated toxicity while maintaining anti-tumor efficacy

  • Variants with altered receptor binding kinetics (faster off-rates) that limit systemic exposure

Delivery innovations:

  • Tumor-targeted IL-12 variants conjugated to tumor-specific antibodies

  • Site-specific administration strategies (intratumoral, intraperitoneal) showing improved efficacy/toxicity ratios

  • Intravenous administration demonstrating superior response rates (40%) compared to subcutaneous delivery (7%) in lymphoma patients

Combination approaches:

  • IL-12 with checkpoint inhibitors to enhance T cell responses

  • Sequential administration protocols to mitigate cytokine release syndrome

  • Co-administration with agents that selectively block toxicity pathways

Production advancements:

  • Endotoxin-free, animal component-free production systems

  • Consistent glycosylation patterns for reduced immunogenicity

  • Stable formulations with extended shelf-life

These approaches collectively aim to harness IL-12's potent anti-tumor properties while addressing the dose-limiting toxicities that have historically restricted its clinical application. Computational modeling combined with experimental validation has proven particularly effective for designing IL-12 variants with improved therapeutic profiles .

What are the methodological challenges in studying IL-12 signaling pathway interactions with other cytokine networks?

Investigating IL-12 signaling interactions with other cytokine networks presents several methodological challenges:

Complex cytokine cascade monitoring:

  • IL-12 administration induces multiple secondary cytokines (IFNγ, TNFα, IP-10, MIG, IL-10, IL-8, VEGF)

  • Researchers need multiplex approaches to simultaneously track these diverse mediators

  • Temporal dynamics vary by cytokine, requiring time-course sampling strategies

Cell-specific response heterogeneity:

  • IL-12 receptors are expressed on NK cells, T cells, and dendritic cells

  • Each cell type shows distinct signaling patterns requiring multiparametric analysis

  • Janus kinase associations differ between receptor components (IL-12Rβ1 with Tyk2; IL-12Rβ2 with Jak2)

Pathway cross-talk visualization:

  • STAT activation by IL-12 involves multiple family members (STAT1, 3, 4, 5) with overlapping functions

  • Requires phospho-flow cytometry or high-resolution signaling protein analysis

  • Integration with other pathways (TCR signaling, other cytokine receptors) adds complexity

In vivo correlation challenges:

  • Cytokine networks in tissue microenvironments differ from in vitro systems

  • Local concentration gradients affect cellular responses

  • Models must account for regulatory feedback mechanisms

Methodological approaches:

  • Single-cell analysis technologies to capture cell-specific responses

  • Systems biology modeling of integrated cytokine networks

  • Ex vivo tissue culture systems that preserve microenvironmental context

  • Genetic approaches (CRISPR) to precisely manipulate pathway components

Researchers investigating IL-12 signaling should employ multiparametric approaches and consider the dynamic, context-dependent nature of these networks rather than studying IL-12 in isolation .

How can researchers optimize His-tag purification protocols specifically for human IL-12 to maximize heterodimer yield?

Optimizing His-tag purification for human IL-12 requires specialized approaches to maintain heterodimer integrity:

Lysis and extraction optimization:

  • Use gentle lysis buffers (avoid harsh detergents that might dissociate the heterodimer)

  • Include protease inhibitors to prevent subunit degradation

  • Maintain reducing conditions to preserve disulfide bonds

  • Consider adding low concentrations of stabilizing agents (10% glycerol, 1mM EDTA)

Immobilized metal affinity chromatography (IMAC) refinements:

  • Nickel or cobalt resins often provide better selectivity than copper

  • Test both N-terminal and C-terminal His-tagged constructs for optimal heterodimer purification

  • Optimize imidazole concentration in wash buffers to remove contaminants without heterodimer loss

  • Consider mild elution conditions using pH gradients rather than high imidazole concentrations

Polishing steps for high purity:

  • Size exclusion chromatography to separate heterodimer (~57 kDa) from free p40 (~42 kDa) and aggregates

  • Ion exchange chromatography exploiting the differing pI values of heterodimer versus individual subunits

  • Affinity chromatography using IL-12 receptor components or specific antibodies

Quality control benchmarks:

  • Final purity should exceed 95% as assessed by SDS-PAGE

  • Endotoxin levels must be <1 EU/μg for research applications

  • Activity verification using receptor binding assays (ED₅₀ of 3-30 ng/mL)

For co-expression systems, optimizing the ratio of p35 to p40 expression can significantly improve heterodimer yield, as p40 is naturally produced in excess and can form homodimers that complicate purification .

What are the most effective approaches for detecting conformational changes in His-tagged IL-12 that might affect biological activity?

Multiple complementary techniques can detect conformational changes in His-tagged IL-12 that impact activity:

Biophysical characterization methods:

  • Circular dichroism (CD) spectroscopy: Monitors secondary structure changes in the heterodimer

  • Differential scanning calorimetry (DSC): Determines thermal stability and folding state

  • Dynamic light scattering (DLS): Detects aggregation or significant conformational changes

  • Intrinsic fluorescence spectroscopy: Reveals tertiary structure alterations through tryptophan exposure

Functional binding assessment:

  • Surface plasmon resonance (SPR) to measure binding kinetics to both receptor subunits

  • Bio-layer interferometry for real-time binding analysis

  • ELISA-based binding assays with conformational antibodies

Stability indicators:

  • SDS-PAGE under non-reducing conditions to verify intact disulfide bridges

  • Native PAGE to assess heterodimer integrity and conformational homogeneity

  • Size exclusion chromatography to detect subtle changes in hydrodynamic radius

Activity correlation studies:

  • Statistical correlation between biophysical parameters and biological activity

  • Accelerated stability studies under various storage conditions

  • Assessment of glycosylation patterns using lectin binding or mass spectrometry

For reliable detection of activity-affecting conformational changes, researchers should establish a baseline characterization of freshly prepared material and use it as a reference for subsequent comparisons. The acidic profile differences observed between secreted and intracellular p35 subunits indicate that post-translational modifications during secretion are critical for proper IL-12 conformation and function .

Product Science Overview

Introduction

Interleukin 12 (IL-12) is a cytokine that plays a crucial role in the immune system by influencing the activities of T cells and natural killer (NK) cells. It is a heterodimeric protein composed of two subunits: IL-12A (p35) and IL-12B (p40). The recombinant form of IL-12, tagged with a histidine (His) tag, is commonly used in research to study its biological functions and potential therapeutic applications.

Structure and Composition

IL-12 is a disulfide-linked heterodimer consisting of two subunits:

  • IL-12A (p35): This subunit has a molecular weight of approximately 23.3 kDa and consists of 203 amino acids .
  • IL-12B (p40): This subunit has a molecular weight of approximately 34.6 kDa and consists of 306 amino acids .

The recombinant IL-12 protein is often produced in various expression systems, such as E. coli or baculovirus-infected insect cells, and is tagged with a hexahistidine (His) tag at the N-terminus or C-terminus to facilitate purification and detection .

Biological Functions

IL-12 is a key cytokine in the immune response, with several important functions:

  1. Activation of T Cells and NK Cells: IL-12 acts as a growth factor for activated T cells and NK cells, enhancing their proliferation and cytotoxic activities .
  2. Induction of IFN-γ Production: IL-12 stimulates the production of interferon-gamma (IFN-γ) by resting peripheral blood mononuclear cells (PBMCs), which is crucial for the immune response against intracellular pathogens .
  3. Th1 Cell Differentiation: IL-12 is essential for the differentiation of naive T cells into Th1 cells, which are involved in the defense against intracellular pathogens and in the pathogenesis of autoimmune diseases .
Applications in Research

Recombinant IL-12 with a His tag is widely used in various research applications:

  • Immunological Assays: It serves as a positive control in assays such as Western blotting and enzyme-linked immunosorbent assays (ELISA) .
  • Functional Studies: Researchers use recombinant IL-12 to study its effects on immune cells and to investigate its potential therapeutic applications in diseases such as cancer and autoimmune disorders .
  • Protein-Protein Interactions: The His tag allows for easy purification and detection of IL-12, facilitating studies on its interactions with other proteins and receptors .

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.