KGF 2 Mouse

Keratinocyte Growth Factor-2 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

KGF 2 Mouse Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 173 amino acids and having a molecular mass of 19.5kDa.
The KGF 2 is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Keratinocyte growth factor 2 (KGF-2), also known as fibroblast growth factor 10 (FGF-10), is a member of the fibroblast growth factor (FGF) family. FGFs are signaling molecules with diverse roles in cell growth, survival, and differentiation. KGF-2 specifically stimulates the proliferation and differentiation of epithelial cells, particularly keratinocytes in the skin. It plays a crucial role in wound healing, embryonic development, and tissue homeostasis.
Description
Recombinant mouse KGF-2 is a single, non-glycosylated polypeptide chain produced in E. coli. It contains 173 amino acids and has a molecular weight of 19.5 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
Sterile white lyophilized (freeze-dried) powder.
Formulation
The protein is lyophilized from a 0.2 µm filtered solution in 1x phosphate-buffered saline (PBS), pH 7.4 containing 5% trehalose.
Solubility
To reconstitute lyophilized mouse KGF-2, it is recommended to dissolve it in sterile 18 MΩ-cm H₂O to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized KGF-2 is stable at room temperature for up to 3 weeks. For long-term storage, it is recommended to store the desiccated protein below -18°C. Upon reconstitution, KGF-2 should be stored at 4°C for 2-7 days or below -18°C for longer periods. The addition of a carrier protein such as 0.1% HSA or BSA is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 97.0%, as determined by reverse-phase high-performance liquid chromatography (RP-HPLC) and sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE).
Biological Activity
The biological activity of the recombinant mouse KGF-2 is comparable to the standard. The ED50, measured by the dose-dependent stimulation of thymidine uptake in BaF3 cells expressing FGF receptors, is less than 0.5 ng/ml.
Synonyms
FGFA, FGF10, FGF-10, KGF-2, Fibroblast growth factor 10.
Source
Escherichia Coli.
Amino Acid Sequence
QALGQDMVSQ EATNCSSSSS SFSSPSSAGR HVRSYNHLQG DVRWRRLFSF TKYFLTIEKN GKVSGTKNED CPYSVLEITS VEIGVVAVKA INSNYYLAMN KKGKLYGSKE FNNDCKLKER IEENGYNTYA SFNWQHNGRQ MYVALNGKGA PRRGQKTRRK NTSAHFLPMT IQT

Q&A

What is KGF and how does it relate to other fibroblast growth factors in mouse models?

Keratinocyte Growth Factor (KGF), also known as Fibroblast Growth Factor 7 (FGF-7), is a cytokine with wide-ranging biological activity. It belongs to the FGF family, which includes other members like FGF-10 (sometimes referred to as KGF-2). KGF is primarily derived from mesenchymal stem cells (MSCs) and plays critical roles in the repair of various organs including the gastrointestinal tract, thymus, lung, and kidney . In mouse models, KGF has demonstrated significant epithelial repair capacity, promoting epithelial cell proliferation and differentiation. While FGF-7 and FGF-10 share structural and functional similarities, FGF-10 (approximately 20 kDa) contains a distinctive serine-rich region near its N-terminus in mouse models .

Methodologically, researchers should note that when designing experiments, the specific selection between KGF/FGF-7 and FGF-10 should be guided by the target tissue and research question, as their receptor binding affinity and downstream signaling pathways exhibit subtle but important differences that may impact experimental outcomes.

How do KGF-modified MSCs differ from unmodified MSCs in experimental applications?

KGF-modified MSCs demonstrate significantly enhanced therapeutic potential compared to unmodified MSCs across multiple experimental models. Studies have shown that lentivirus-mediated KGF gene introduction into rat bone marrow MSCs results in significantly higher KGF mRNA and protein expression levels (2.323±0.812 vs. 0.824±0.313 for mRNA; 2.585±0.883 vs. 0.865±0.462 for protein) . This overexpression translates to functional differences in tissue repair capabilities.

In ulcerative colitis (UC) rat models, for example, KGF-modified MSCs (MSCs-KGF) showed superior efficacy in reducing Disease Activity Index (DAI), Colon Macroscopic Damage Index (CMDI), and Tissue Damage Index (TDI) scores compared to both unmodified MSCs and MSCs carrying empty vectors (MSCs-vec) . The enhanced therapeutic effect appears to be mediated through multiple mechanisms:

  • Increased expression of tight junction proteins (e.g., claudin-1)

  • Enhanced proliferation markers (PCNA)

  • Greater anti-inflammatory effects (reduced TNF-α, IL-6, IL-8)

  • Increased immunomodulatory cytokine production (IL-10)

  • Enhanced activation of the PI3K/Akt pathway

  • More effective inhibition of NF-κB nuclear translocation

When designing experiments with modified versus unmodified MSCs, researchers should account for these mechanistic differences and consider appropriate readouts to capture the multifaceted effects of KGF modification.

What are the optimal methods for genetically modifying MSCs to express KGF for mouse model experiments?

Based on established protocols, lentiviral vector transduction represents the most efficient method for introducing the KGF gene into mouse MSCs. The optimal methodology includes:

  • MSC preparation: Isolate and expand bone marrow MSCs to the 4th generation in standard culture conditions

  • Vector preparation: Package KGF recombinant adenovirus with appropriate promoters

  • Infection protocol: Calculate virus amount according to a multiplicity of infection (MOI) of 100 after 24 hours of MSC culture

  • Controls: Always include unmodified MSCs and MSCs infected with empty vectors (MSCs-vec) as essential experimental controls

  • Validation: Confirm KGF expression using both RT-PCR and Western blot analysis to verify both transcriptional and translational success

  • Functional testing: Assess biological activity of the expressed KGF through in vitro assays before proceeding to in vivo applications

For optimal transduction efficiency, seeding MSCs at 70-80% confluence before viral infection is recommended. Post-infection, researchers should allow 48-72 hours for KGF expression to stabilize before conducting validation assays or using the cells for experimental purposes. The choice between adenoviral and lentiviral vectors depends on whether transient or stable expression is desired, with lentiviral systems preferred for long-term studies.

What in vivo delivery methods are most effective for administering KGF-modified MSCs in mouse disease models?

The effectiveness of KGF-modified MSC delivery varies by disease model and research objective. Based on comparative studies, several administration routes have demonstrated efficacy:

  • Tail vein injection: Most commonly used for systemic delivery, this method shows good engraftment in UC models and allows MSCs to traffic to sites of inflammation . This approach requires careful cell dose calculation, typically 1-5×10^6 cells per adult mouse.

  • Direct tissue injection: For localized tissue repair (e.g., spinal cord injury), direct administration provides higher local concentrations but requires surgical expertise.

  • Intraperitoneal injection: Alternative systemic delivery route that may offer different biodistribution profiles compared to intravenous administration.

  • Encapsulated delivery systems: For sustained release applications, biomaterial encapsulation can enhance MSC survival and extend KGF secretion duration.

Key methodological considerations include:

  • Cell preparation: Fresh MSCs-KGF preparations show higher viability and functionality

  • Pre-conditioning: Hypoxic pre-conditioning (2-5% O₂ for 24h) may enhance survival and therapeutic effect

  • Tracking methods: Labeling cells with fluorescent dyes or using sex-mismatched transplantation (male MSCs into female recipients) enables tracking of cell fate

  • Timing: Administration during acute phase of injury/inflammation typically shows better therapeutic outcomes than during chronic stages

The selection of delivery method should be guided by the specific research question, target tissue accessibility, and desired duration of therapeutic effect.

How do KGF-modified MSCs modulate PI3K/Akt and NF-κB signaling pathways in inflammatory disease models?

KGF-modified MSCs exert their therapeutic effects through sophisticated modulation of key cellular signaling pathways. In UC models, MSCs-KGF demonstrate significantly enhanced regulatory effects on the PI3K/Akt and NF-κB signaling axes compared to unmodified MSCs .

The mechanistic cascade appears to proceed as follows:

  • Enhanced PI3K/Akt activation: MSCs-KGF significantly increase phosphorylation of PI3K and Akt in intestinal tissues compared to MSCs or MSCs-vec. This activation is quantifiable through Western blot analysis of phosphorylated:total protein ratios.

  • Inhibition of NF-κB nuclear translocation: KGF overexpression in MSCs results in marked reduction of NF-κB p65 nuclear translocation in colonic tissues of UC rats. This effect correlates with reduced inflammatory cytokine production.

  • Bidirectional regulation of inflammatory mediators: MSCs-KGF administration leads to significant decreases in pro-inflammatory cytokines (TNF-α, IL-6, IL-8) while simultaneously increasing anti-inflammatory IL-10 levels in intestinal tissues .

  • Enhancement of epithelial barrier function: Activation of PI3K/Akt pathway by MSCs-KGF upregulates tight junction proteins like claudin-1, reinforcing epithelial barrier integrity.

Methodologically, researchers investigating these pathways should employ multiple complementary techniques:

  • Phospho-specific Western blotting for PI3K/Akt activation status

  • Nuclear/cytoplasmic fractionation to assess NF-κB translocation

  • Immunohistochemistry to visualize pathway activation in tissue context

  • Selective pathway inhibitors (e.g., wortmannin for PI3K) to confirm causality

  • Multiplex cytokine assays to comprehensively assess inflammatory profile changes

Understanding these signaling interactions is crucial for optimizing MSCs-KGF therapy and potentially developing small molecule approaches that might mimic these beneficial effects.

What are the comparative effects of FGF-7 versus FGF-10 in mouse organ development and tissue repair models?

FGF-7 (KGF) and FGF-10 demonstrate distinct yet overlapping roles in organ development and tissue repair in mouse models, with important implications for experimental design:

ParameterFGF-7 (KGF)FGF-10Methodological Considerations
Receptor specificityPrimarily FGFR2bFGFR2b and FGFR1bUse receptor-specific antagonists to distinguish pathway contributions
Structural features~19 kDa protein~20 kDa with serine-rich N-terminus Protein tag position may affect biological activity
Lung development/repairModerate effect on alveolar epitheliumCritical for branching morphogenesis and alveolar cell fate selection Timing of administration affects developmental outcomes
Salivary gland biologySupports epithelial repairEssential for duct homeostasis and acinar cell differentiation Analyze both structural and functional outcomes
Intestinal epitheliumStrong proliferative effect on intestinal epithelial cells Moderate effect on intestinal repairUse BrdU/EdU pulse-chase to quantify proliferation rates
Pancreatic developmentLimited effectCritical for pancreatic bud outgrowth Examine both endocrine and exocrine compartments

For experimental applications, researchers should note that:

  • Dosage requirements differ significantly (typically 10-50 ng/mL for in vitro studies)

  • Temporal expression patterns during development are non-redundant

  • FGF-10 demonstrates stronger effects in certain stem cell differentiation protocols

  • Combination treatments may yield synergistic effects in some repair models

When designing comparative studies, it's essential to control for protein stability differences and to assess multiple timepoints, as the kinetics of response may vary between these growth factors.

How can researchers address variability in KGF expression levels after lentiviral transduction of MSCs?

Variability in KGF expression following lentiviral transduction represents a common technical challenge that can impact experimental reproducibility. To address this issue, researchers should implement the following methodological approaches:

  • Standardize MOI calculation: Precisely quantify viral titers using qPCR-based methods rather than relying solely on fluorescent reporter expression. For KGF lentiviral constructs, an MOI of 100 has been demonstrated to provide optimal transduction efficiency without compromising MSC viability .

  • Implement cell sorting strategies: After transduction, consider using fluorescence-activated cell sorting (FACS) to select for cells with consistent transgene expression levels. This approach can reduce heterogeneity in the MSC-KGF population.

  • Establish expression verification protocols: Develop robust quantitative RT-PCR and Western blot protocols with appropriate housekeeping controls. Expected KGF mRNA expression in successfully transduced MSCs should be approximately 2.3-fold higher than baseline MSCs, while protein expression typically increases approximately 2.5-fold .

  • Monitor expression stability: Assess KGF expression over multiple passages to ensure stability of the genetic modification. Expression typically begins to decline after 8-10 passages.

  • Address clonal variation: Generate and characterize multiple MSC-KGF clones, selecting those with consistent expression profiles for experimental use.

  • Optimize culture conditions: Standardize serum lots, passage numbers, and confluence levels at transduction to minimize variability in transduction efficiency.

For validation purposes, researchers should confirm that KGF secreted by modified MSCs remains biologically active using appropriate bioassays, such as epithelial cell proliferation assays, before proceeding to in vivo applications.

What are the best methods for detecting KGF protein expression in target tissues after MSC transplantation?

Detecting KGF protein expression in target tissues following MSC transplantation requires a strategic combination of complementary techniques to overcome sensitivity and specificity challenges:

  • Immunohistochemistry (IHC): This remains the gold standard for localizing KGF protein within tissue microenvironments. For optimal results:

    • Use antigen retrieval methods (citrate buffer pH 6.0)

    • Employ tyramide signal amplification for low-abundance detection

    • Include positive controls (direct KGF injection) and negative controls (isotype antibodies)

    • Consider dual staining with MSC markers to confirm cellular source

  • Western blot analysis: Quantitative assessment of total KGF protein levels in tissue homogenates provides complementary data to IHC:

    • Expected fold-change in KGF protein expression in MSCs-KGF treated tissues is 1.121±0.379 compared to 0.010±0.010 in control tissues

    • Use gradient gels (10-20%) for optimal separation of the approximately 19-20 kDa KGF protein

    • Consider membrane fractionation to enhance detection of membrane-associated KGF

  • qRT-PCR with species-specific primers: When using male-derived MSCs in female recipients, species-specific or sex chromosome-specific primers can distinguish transplanted-cell derived KGF expression from endogenous expression .

  • ELISA-based techniques: For quantification of secreted KGF in tissue fluid or serum:

    • Commercial ELISA kits typically have detection limits ~10 pg/mL

    • Consider concentration steps for samples with low KGF levels

    • Be aware of potential cross-reactivity with other FGF family members

  • Mass spectrometry approaches: For unambiguous identification of KGF and potential post-translational modifications:

    • Selected reaction monitoring (SRM) offers enhanced sensitivity

    • Requires careful sample preparation to enrich low-abundance proteins

Temporal considerations are crucial, as peak KGF expression in target tissues typically occurs 3-7 days post-MSC transplantation, with significant decrease by 14 days in most models.

How do KGF and related growth factors influence stem cell differentiation pathways in organoid models?

KGF (FGF-7) and related growth factors like FGF-10 play sophisticated roles in directing stem cell fate decisions in organoid culture systems, with applications spanning multiple organ systems:

  • Intestinal organoids: KGF promotes intestinal stem cell proliferation and drives differentiation toward absorptive enterocyte lineage. Methodologically, KGF supplementation (50 ng/mL) enhances organoid formation efficiency approximately 1.8-fold compared to standard conditions . Timing of KGF addition is critical - early administration (days 0-3) promotes stem cell expansion, while later addition (days 4-7) enhances differentiation.

  • Lung organoids: FGF-10 is particularly important for alveolar cell fate selection in lung organoid models, where it directs the maintenance of alveolar type 2 (AT2) cells . For optimal results, pulsed rather than continuous FGF-10 exposure (alternating 24-hour treatment/withdrawal cycles) better recapitulates developmental signaling.

  • Pancreatic organoids: FGF-10 supplementation is crucial for organotypic pancreatic organoid development, particularly for establishing proper epithelial-mesenchymal interactions . Concentration-dependent effects are observed: low doses (10 ng/mL) maintain progenitor state while higher doses (50 ng/mL) promote differentiation toward exocrine lineages.

  • Salivary gland organoids: Both FGF-7 and FGF-10 contribute to salivary gland development, with FGF-10 particularly important for duct homeostasis and acinar cell differentiation . For methodology optimization, combining FGF-10 with basement membrane components enhances glandular structure formation.

  • Gastric organoids: FGF-10 promotes gastric fundus specification in both mouse and human organoid models . The temporal sequence of growth factor exposure is critical - initial WNT activation followed by FGF-10 treatment yields optimal fundic specification.

For organoid applications, researchers should consider:

  • Growth factor stability (typically 48-72 hours at 37°C)

  • Matrix composition interactions (Matrigel vs. synthetic alternatives)

  • Species-specific response differences

  • Combinatorial effects with other niche factors

What are the latest findings on KGF/FGF signaling crosstalk with other pathways in tissue regeneration models?

Recent research has unveiled complex signaling interactions between KGF/FGF pathways and other major signaling networks in tissue regeneration, with important implications for therapeutic targeting:

  • PI3K/Akt pathway crosstalk: KGF-modified MSCs significantly enhance phosphorylation of PI3K and Akt in target tissues, creating a pro-regenerative environment . This activation appears bidirectional - PI3K inhibition reduces KGF expression, suggesting a feed-forward amplification loop that researchers can leverage by combining KGF therapy with selective PI3K activators.

  • NF-κB signaling intersection: KGF-mediated inhibition of NF-κB nuclear translocation represents a primary anti-inflammatory mechanism . Experimentally, this interaction can be monitored through subcellular fractionation and Western blotting for phosphorylated IκB. The timing of this inhibition is crucial - effects peak at 24-48 hours post-treatment.

  • Wnt/β-catenin coordination: Emerging evidence indicates that FGF-10 works cooperatively with Wnt/β-catenin signaling in gastric fundus specification and tissue patterning . This relationship appears tissue-specific, as antagonistic interactions are observed in other contexts like pancreatic development. Researchers should carefully track both pathway activities when manipulating either system.

  • Hedgehog pathway interactions: FGF signaling modulates Sufu- and Spop-mediated downregulation of Hedgehog signaling in pancreatic β cell differentiation . This intricate relationship requires sophisticated experimental tracking, ideally using reporter constructs for both pathways.

  • MAPK/ERK pathway convergence: FGF-10 influences MAPK/ERK signaling to regulate squamous-columnar junction specification and salivary gland development. This interaction demonstrates dose-dependent characteristics, with high FGF concentrations activating sustained ERK phosphorylation while lower doses trigger transient responses.

For investigating these complex signaling networks, researchers should implement:

  • Multiplexed phosphoprotein analysis

  • Selective pathway inhibitors used in combination

  • Temporal profiling to capture dynamic relationships

  • Single-cell approaches to address cellular heterogeneity

  • Computational modeling to predict intervention points

Understanding these pathway interactions will facilitate the development of more targeted approaches to enhance tissue regeneration across multiple organ systems.

How should researchers interpret contradictory findings regarding KGF efficacy across different mouse disease models?

When confronting contradictory findings regarding KGF efficacy across mouse disease models, researchers should implement a systematic analytical approach that considers multiple variables:

  • Disease model heterogeneity: The TNBS-induced UC model shows consistent response to KGF-modified MSCs , while other inflammatory models may demonstrate variable outcomes. Critical methodological differences include:

    • Induction method (chemical vs. genetic vs. immunological)

    • Disease severity at treatment initiation

    • Route of administration (systemic vs. local)

    • Treatment duration and dosing schedule

  • Strain-dependent responses: Different mouse strains exhibit variable baseline FGFR expression profiles and inflammatory responses. For experimental design:

    • Always report complete strain information

    • Consider testing multiple strains when contradictory literature exists

    • Account for sex-based differences in KGF response

  • KGF source and preparation variables:

    • Recombinant KGF vs. cell-secreted KGF may show different bioavailability profiles

    • KGF from different species (human vs. mouse) has different receptor binding characteristics

    • Storage conditions and freeze-thaw cycles affect protein activity

  • Endpoint selection and timing:

    • Acute vs. chronic models may show different optimal treatment windows

    • Proliferation markers (PCNA) may show positive response while functional outcomes remain unchanged

    • Histological improvement may precede functional recovery

When analyzing contradictory findings, researchers should construct a comprehensive comparison table documenting:

  • Model parameters (strain, age, induction method)

  • Treatment details (dose, route, timing)

  • Assessment methods (histological, molecular, functional)

  • Statistical approaches (sample size, analysis methods)

This structured analysis often reveals that apparent contradictions result from methodological differences rather than true biological inconsistencies in KGF action.

What statistical approaches are most appropriate for analyzing the efficacy of KGF-modified MSCs in mouse disease models?

Selecting appropriate statistical methods for analyzing KGF-modified MSC efficacy requires careful consideration of experimental design and data characteristics:

  • Study design considerations:

    • For multiple treatment groups (control, challenged control, MSCs, MSCs-vec, MSCs-KGF), one-way ANOVA with appropriate post-hoc tests (Tukey or Bonferroni) is recommended for continuous variables

    • For longitudinal assessments (disease progression over time), repeated measures ANOVA or mixed-effects models provide superior analysis

    • For survival outcomes, Kaplan-Meier analysis with log-rank tests is appropriate

  • Sample size determination:

    • Power analysis should account for expected effect size differences between unmodified MSCs and KGF-MSCs

    • Based on published data, detecting significant differences in DAI, CMDI, and TDI scores requires minimum n=6-8 per group

    • For more subtle molecular endpoints (e.g., signaling pathway activation), larger sample sizes (n=10-12) may be necessary

  • Handling non-normal distributions:

    • Histological scoring data often follows non-normal distribution and should be analyzed using non-parametric methods (Kruskal-Wallis with Dunn's post-hoc)

    • Consider data transformations when appropriate (log transformation for cytokine concentrations)

    • Report median and interquartile range rather than mean±SD for non-normal data

  • Multivariate approaches:

    • Principal component analysis can help integrate multiple outcome measures

    • Partial least squares discriminant analysis may identify patterns distinguishing responders from non-responders

    • Hierarchical clustering of transcriptomic or proteomic data can reveal mechanism-based groupings

  • Correlation analysis:

    • Pearson or Spearman correlation between KGF expression levels and outcome measures helps establish dose-response relationships

    • Multiple regression models can identify key predictors of therapeutic response

For transparent reporting, researchers should:

  • Predefine primary and secondary endpoints

  • Account for multiple comparisons

  • Report effect sizes alongside p-values

  • Consider blinded assessment of subjective endpoints

  • Provide complete dataset accessibility

These statistical approaches enhance reproducibility and facilitate meaningful comparison across different experimental models and therapeutic strategies.

Product Science Overview

Introduction

Keratinocyte Growth Factor-2 (KGF-2), also known as Fibroblast Growth Factor 10 (FGF-10), is a member of the fibroblast growth factor (FGF) family. This family consists of at least 23 related peptides that play crucial roles in various biological processes, including cell growth, development, and tissue repair . KGF-2 is particularly known for its role in stimulating the proliferation and differentiation of epithelial cells, making it a significant factor in wound healing and tissue regeneration .

Structure and Function

KGF-2 is produced by fibroblasts in the dermis, granulation tissue, and intraepithelial cells. It specifically acts on cells of epithelial origin, such as skin keratinocytes and intestinal epithelial cells, by binding to the FGF receptor 2-IIIb (FGFR2-IIIb) . The protein sequence of KGF-2 shows high homology with other members of the FGF family, particularly FGF-7 (KGF-1), indicating similar biological functions .

Biological Role

KGF-2 plays a pivotal role in various biological processes:

  • Wound Healing: KGF-2 is involved in the repair of cutaneous wounds by stimulating keratinocyte migration and proliferation. It is rapidly induced after skin injury and contributes to the re-epithelialization process .
  • Hair Growth: KGF-2 is found in dermal papilla fibroblasts and its receptor, FGFR2, is present in the outer root sheath of keratinocytes. This interaction promotes hair growth by stimulating the proliferation of hair follicle cells .
  • Gastrointestinal Epithelium Repair: KGF-2 enhances the healing of mucosal wounds in the gastrointestinal tract by promoting the growth and migration of intestinal epithelial cells .
Recombinant Production

The recombinant production of KGF-2 involves the expression and purification of the protein using various biotechnological techniques. One common method is the fusion of KGF-2 with a small ubiquitin-related modifier (SUMO) to enhance protein expression and prevent degradation . The fusion protein is then purified using affinity chromatography, and the SUMO tag is removed to obtain the recombinant KGF-2 .

Applications

KGF-2 has several potential applications in medical and cosmetic fields:

  • Therapeutic Use: Due to its role in wound healing and tissue regeneration, KGF-2 is being explored as a therapeutic agent for treating chronic wounds, burns, and other skin injuries .
  • Cosmetic Use: KGF-2 is also being investigated for its potential in promoting hair growth and improving skin health, making it a valuable ingredient in cosmetic products .

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.