PKM2 Mouse

Tumour Type M2 Pyruvate Kinase Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Genetic Engineering Approaches

PKM2 mouse models employ tissue-specific or germline knockout strategies:

Model TypeGenetic ModificationKey FeaturesSources
Germline PKM2 knockoutPkm2−/− (global deletion)Systemic PKM2 loss; spontaneous HCCs in aging
Tissue-specific knockoutConditional deletion (e.g., β-cell, myeloid)Cell-type-specific PKM2 loss
PKM2 aggregation modelsOverexpression of mutant PKM2 (e.g., ΔA2)Accelerated senescence/aging phenotypes

Key Insights:

  • Compensation mechanisms: PKM1 isoform upregulation occurs in PKM2 knockout models, preserving partial metabolic function .

  • Isoform-specific effects: PKM2 deletion in β-cells does not impair glucose tolerance, while germline deletion causes metabolic distress .

Cancer Biology

PKM2 drives glycolysis and tumor metabolism. Studies using PKM2-silenced xenograft models reveal:

ParameterPKM2-Silenced TumorsControl Tumorsp-valueSource
Tumor growth rateSlowerFaster<0.05
Glycolytic fluxReducedNormal
PD-L1 expressionDownregulatedUpregulated

Mechanisms:

  • PKM2 depletion suppresses the Warburg effect, reducing lactate production and nucleotide synthesis .

  • PKM2 inhibition enhances anti-tumor immunity by modulating PD-L1 expression and CD8+ T-cell activity .

Type 1 Diabetes (T1D)

PKM2 knockout in β-cells improves glucose homeostasis:

MetricPKM2-Knockout MiceWild-Type MiceSource
Blood glucose (mmol/L)12.5 ± 1.218.3 ± 1.5
β-cell apoptosisReducedHigh
Oxidative stress (ROS)LowerElevated

Mechanisms:

  • PKM2 deletion reduces β-cell injury by dampening oxidative stress and inflammatory cytokines (e.g., IL-6, TNF-α) .

Aging and Senescence

PKM2 aggregation drives age-related metabolic dysfunction:

TreatmentLifespan (Weeks)Senescence MarkersSource
Untreated80 ± 5High SASP, SA-β-gal
K35/K27 (PKM2 disaggregators)95 ± 3Reduced SASP, p21

Mechanisms:

  • PKM2 aggregates impair glycolytic flux, promoting senescence-associated secretory phenotype (SASP) .

  • Small molecules (K35/K27) dissolve aggregates, restoring metabolic function and extending lifespan .

Cardiovascular Diseases

Myeloid-specific PKM2 deletion reduces atherosclerosis:

ParameterPKM2 mye-KO MiceControl MiceSource
Aortic lesion area (%)30 ± 555 ± 6
Macrophage efferocytosisEnhancedImpaired
LRP-1 expressionUpregulatedNormal

Mechanisms:

  • PKM2 deletion shifts macrophages to anti-inflammatory (M2) polarization, reducing foam cell formation .

Therapeutic Implications

PKM2 targeting strategies include:

  1. Activators/Inhibitors:

    • PKM2 activators (e.g., PKA) enhance glycolysis, improving insulin secretion in β-cells .

    • PKM2 inhibitors suppress tumor growth but require optimization for in vivo efficacy .

  2. Aggregate Disruptors:

    • K35/K27 extend lifespan in aging models by resolving PKM2 aggregates .

Challenges and Future Directions

  • Tissue specificity: Off-target effects limit germline knockout models; conditional models are preferred .

  • Species differences: PKM2 isoform dominance varies between mice and humans, necessitating cross-validation .

  • Clinical translation: PKM2-targeted therapies require biomarker-driven patient stratification to address heterogeneous responses .

Product Specs

Introduction
Pyruvate kinase is a crucial enzyme in the glycolytic pathway. The M2 isoenzyme of pyruvate kinase exhibits high expression levels in tumor cells and can be quantified in the plasma of patients diagnosed with advanced breast cancer. This marker proves valuable in monitoring disease activity, assessing chemotherapy sensitivity, and detecting recurrence.
Description
Recombinant PKM2 Mouse protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 554 amino acids (1-531 a.a) with a molecular weight of 60.2kDa. A 23 amino acid His-tag is fused to the N-terminus of PKM2, and the protein undergoes purification using proprietary chromatographic methods.
Physical Appearance
Clear solution subjected to sterile filtration.
Formulation
PKM2 protein solution at a concentration of 0.5mg/ml. The solution contains 20mM Tris-HCl (pH 8.5), 0.2M NaCl, 1mM DTT, and 30% glycerol.
Stability
For short-term storage (2-4 weeks), maintain at 4°C. For extended periods, store frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 85% as assessed by SDS-PAGE.
Biological Activity
Specific activity: greater than 500 units/mg. One unit is defined as the amount of enzyme required to catalyze the conversion of 1 picomole of phospho(enol)pyruvate to pyruvate per minute at pH 7.5 and a temperature of 37°C.
Synonyms

Pyruvate kinase isozymes M1/M2, EC 2.7.1.40, Pyruvate kinase muscle isozyme, Pyruvate kinase 2/3, Cytosolic thyroid hormone-binding protein, CTHBP, THBP1, M2PK, PKM2, PK3, PK2, PKM, TCB, OIP3, MGC3932, Tumor Type M2 Pyruvate Kinase.

Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MGSMPKPHSE AGTAFIQTQQ LHAAMADTFL EHMCRLDIDS APITARNTGI ICTIGPASRS VEMLKEMIKS GMNVARLNFS HGTHEYHAET IKNVREATES FASDPILYRP VAVALDTKGP EIRTGLIKGS GTAEVELKKG ATLKITLDNA YMEKCDENIL WLDYKNICKV VEVGSKIYVD DGLISLQVKE KGADFLVTEV ENGGSLGSKK GVNLPGAAVD LPAVSEKDIQ DLKFGVEQDV DMVFASFIRK AADVHEVRKV LGEKGKNIKI ISKIENHEGV RRFDEILEAS DGIMVARGDL GIEIPAEKVF LAQKMMIGRC NRAGKPVICA TQMLESMIKK PRPTRAEGSD VANAVLDGAD CIMLSGETAK GDYPLEAVRM QHLIAREAEA AIYHLQLFEE LRRLAPITSD PTEAAAVGAV EASFKCCSGA IIVLTKSGRS AHQVARYRPR APIIAVTRNP QTARQAHLYR GIFPVLCKDA VLNAWAEDVD LRVNLAMDVG KARGFFKKGD VVIVLTGWRP GSGFTNTMRV VPVP.

Q&A

What is the expression pattern of PKM2 in normal mouse tissues?

PKM2 demonstrates widespread expression across both mouse and human adult tissues with distinct cell type-specific patterns. Immunohistochemical analysis of wild-type mouse tissues reveals PKM2 expression in several specific cell types, including kidney tubular cells, intestinal epithelial cells, pancreatic islet cells, and lung epithelial cells . Other PKM2-expressing cell types include spermatogonia, adrenal gland cortex cells, and choroid plexus cells in the brain . RNA-sequencing data from multiple mouse tissues confirms that PKM2 is the predominant isoform in many adult tissues, characterized by low inclusion of the PKM1-specific exon 9 . This expression pattern suggests that PKM2 serves non-redundant functions in different cell types.

What compensatory mechanisms occur in PKM2-deficient mice?

In PKM2-deficient mice, a consistent compensatory increase in PKM1 expression occurs in tissues that normally express PKM2. This isoform switching has been documented in multiple studies using different PKM2 knockout models:

  • In germline PKM2-null mice, immunohistochemical analysis revealed PKM1 expression in cell types that normally express PKM2 .

  • Myeloid-specific PKM2 knockout mice show a compensatory increase in PKM1 in myeloid tissues .

  • Quantitative RT-PCR analysis confirms dramatically increased PKM1 transcript levels in PKM2-null tissues relative to wild-type tissues .

Interestingly, despite this compensation, total PKM transcript levels are often reduced in null tissues compared to wild-type, potentially due to the generation of PKMskip transcripts that undergo nonsense-mediated decay . This compensatory upregulation of PKM1 suggests functional redundancy between the isoforms for some but not all biological functions.

What are the different types of PKM2 knockout mouse models available for research?

Researchers have developed several PKM2 knockout mouse models that target specific cell types or tissues:

  • Germline PKM2 knockout mice (Pkm2^-/-): These mice have complete deletion of PKM2 in all tissues and cells, achieved by deleting exon 10 of the PKM gene, which is specific to the PKM2 isoform .

  • Cell type-specific PKM2 knockout models:

    • β-cell-specific PKM2 knockout (PKM2^fl/fl^cre+): Created for studying the role of PKM2 in diabetes, these mice have PKM2 deleted specifically in pancreatic β-cells .

    • Myeloid-specific PKM2 knockout (PKM2^fl/fl^LysMCre+/-): Generated using the LysM-Cre system to delete PKM2 specifically in myeloid cells, including neutrophils and macrophages .

    • Myeloid-specific PKM2 knockout on atherosclerosis-prone background (PKM2^mye-KO^Ldlr^-/-^): These mice combine myeloid-specific PKM2 deletion with LDL receptor deficiency to study the role of PKM2 in atherosclerosis .

Each model provides unique insights into the tissue-specific functions of PKM2 and allows researchers to study the consequences of PKM2 loss in specific disease contexts without the confounding effects of systemic PKM2 deletion.

What validation methods should be used to confirm successful PKM2 knockout in mouse models?

Proper validation of PKM2 knockout is crucial for result interpretation. Based on published research, multiple complementary approaches should be employed:

  • Genomic PCR: Confirm the presence of Cre recombinase genes and/or deletion of targeted PKM2 exons .

  • Western blotting: Verify the absence of PKM2 protein in targeted tissues and the presence of compensatory PKM1 expression. For tissue-specific knockouts, confirm normal PKM2 expression in non-targeted tissues .

  • Immunohistochemistry/Immunofluorescence: Visually confirm the absence of PKM2 in targeted cells while verifying its presence in non-targeted cells .

  • Quantitative RT-PCR (qRT-PCR): Measure PKM2 transcript levels and quantify the compensatory increase in PKM1 transcripts. Also useful for assessing total PKM transcript levels .

  • Functional validation: Measure PKM2-dependent functions, such as lactate production during cellular stimulation, to confirm functional consequences of PKM2 deletion .

For tissue-specific knockouts, it is particularly important to demonstrate specificity by showing normal PKM2 expression in non-targeted tissues alongside complete deletion in targeted cells .

How does PKM2 deletion affect glucose metabolism and diabetes in mouse models?

PKM2 deletion in pancreatic β-cells significantly improves glucose metabolism in Type 1 Diabetes (T1D) mouse models. Studies have shown that:

  • β-cell-specific PKM2 knockout mice with streptozotocin (STZ)-induced T1D exhibit decreased blood glucose levels compared to diabetic mice with intact PKM2 .

  • PKM2 knockout in the diabetic model attenuates β-cell injury by suppressing cell apoptosis .

  • The protective effects of PKM2 deletion appear to be mediated through:

    • Reduction in oxidative stress markers in pancreatic tissues

    • Suppression of inflammatory responses, as evidenced by decreased serum levels of pro-inflammatory cytokines (IL-6 and TNF-α)

  • Glucose tolerance tests and insulin tolerance tests demonstrate improved ability to catabolize glucose in diabetic mice lacking PKM2 .

These findings suggest that PKM2 inhibition could represent a novel therapeutic approach for T1D by preserving β-cell function and reducing hyperglycemia through attenuating oxidative stress and inflammation.

What is the role of PKM2 in inflammation and acute lung injury in mouse models?

Myeloid-specific PKM2 deletion dramatically reduces inflammatory responses and protects against acute lung injury (ALI) in mouse models. The evidence includes:

  • PKM2-deficient polymorphonuclear neutrophils (PMNs) show significantly impaired inflammatory functions:

    • Reduced capacity for fMLP- or PMA-induced degranulation of secondary and tertiary granules

    • Decreased reactive oxygen species (ROS) production

    • Impaired transfilter migration and infiltration in response to inflammatory stimuli

  • In lipopolysaccharide (LPS)-induced ALI mouse models, myeloid-specific PKM2 knockout mice exhibited:

    • Less lung tissue damage as shown by histological examination

    • Significantly reduced infiltration of CD11b- and MPO-positive neutrophils in alveolar spaces

    • Lower myeloperoxidase (MPO) activity in lung tissues

    • Fewer neutrophils in bronchoalveolar lavage fluid (BALF)

  • Physiological markers of lung injury were also improved in PKM2-deficient mice:

    • Lower wet/dry weight ratio, indicating reduced pulmonary edema

    • Decreased levels of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) in both serum and BALF

These findings establish PKM2 as a critical regulator of neutrophil-mediated inflammatory responses and suggest that targeting myeloid PKM2 could be a therapeutic strategy for inflammatory conditions like ALI.

What is the relationship between PKM2 deficiency and liver cancer development in aging mice?

PKM2-null mice develop spontaneous hepatocellular carcinomas (HCCs) at high frequency as they age, revealing an unexpected tumor suppressor role of PKM2 in the liver. Key findings include:

  • Aged PKM2-deficient mice (55-94 weeks) display a dramatic incidence of liver tumors not observed in wild-type mice .

  • This hepatic tumorigenesis appears to be preceded by progressive systemic metabolic disturbances, suggesting a causal relationship between metabolic dysregulation and liver cancer development .

  • The PKM2-deficient HCC model recapitulates the correlation between metabolic disease and HCC observed in humans .

  • Analysis of human HCC RNA-seq data and immunohistochemistry on primary human tumor sections revealed heterogeneous PKM2 expression across human HCCs, with some tumors showing very high expression and others showing very low levels .

This apparent paradox—where PKM2 absence promotes cancer in the liver despite PKM2 being highly expressed in many other cancer types—highlights the complex, context-dependent roles of PKM2 in cellular metabolism and tumorigenesis. The PKM2-null mouse represents a valuable model for studying how systemic metabolic aberrations can lead to hepatocellular carcinoma development.

How does myeloid-specific PKM2 deletion affect atherosclerosis progression?

Myeloid cell-specific deletion of PKM2 in atherosclerosis-prone mice leads to significant reduction in atherosclerotic lesion development through multiple mechanisms:

  • PKM2^mye-KO^Ldlr^-/-^ mice fed a Western diet for 14 weeks showed:

    • Reduced atherosclerotic lesions in both whole aorta and aortic sinus

    • Smaller necrotic core area in atherosclerotic plaques

    • These effects occurred despite high cholesterol and triglyceride levels

  • The protective mechanisms include:

    • Decreased macrophage content in lesions

    • Reduced MCP-1 (monocyte chemoattractant protein 1) levels in plasma

    • Impaired transmigration of macrophages in response to MCP-1

    • Decreased glycolytic rate in macrophages

  • Macrophages from myeloid-specific PKM2-deficient mice exhibited:

    • Reduced expression of proinflammatory genes, including MCP-1, IL-1β, and IL-12

    • Enhanced efferocytosis (clearance of apoptotic cells)

    • Upregulation of LRP-1 (LDLR-related protein-1) in macrophages both in vitro and in atherosclerotic lesions in vivo

  • Silencing LRP-1 in PKM2-deficient macrophages restored inflammatory gene expression and reduced efferocytosis, establishing LRP-1 as a mechanistic link between PKM2 deficiency and improved atherosclerosis outcomes .

These findings identify myeloid PKM2 as a potential therapeutic target in atherosclerosis and highlight the importance of cell type-specific metabolic programming in cardiovascular disease.

What are the optimal methods for validating tissue-specific PKM2 expression patterns in mouse models?

Based on published research, a multi-modal approach is recommended for comprehensive validation of PKM2 expression patterns:

  • Immunohistochemistry (IHC):

    • Provides cellular resolution of PKM2 expression within tissues

    • Allows identification of specific PKM2-expressing cell types within heterogeneous tissues

    • Should be performed with isoform-specific antibodies validated for specificity

  • RNA-sequencing analysis:

    • Quantifies PKM splicing patterns at transcriptome level

    • Measures inclusion rates of PKM1-specific exon 9 versus PKM2-specific exon 10

    • Enables comparison between mouse and human tissues

  • Quantitative RT-PCR (qRT-PCR):

    • Provides quantitative assessment of PKM isoform transcript levels

    • Can measure total PKM transcripts alongside isoform-specific expressions

    • Useful for detecting alternative splicing events like PKMskip

  • Western blotting:

    • Confirms protein-level expression of PKM isoforms

    • Can detect compensatory changes in PKM1 expression when PKM2 is deleted

    • Should include controls for antibody specificity

  • Immunofluorescence:

    • Enables co-localization studies with cell type-specific markers

    • Particularly useful for confirming cell-specific knockout in conditional models

Researchers should employ multiple complementary approaches, as each method has distinct strengths and limitations. Comparing results across techniques provides the most comprehensive and reliable assessment of PKM2 expression patterns.

What functional assays should be used to evaluate the metabolic impact of PKM2 deletion in mouse cells?

To comprehensively assess the metabolic consequences of PKM2 deletion, researchers should employ multiple functional assays that evaluate different aspects of cellular metabolism:

These functional assays should be performed alongside appropriate controls and under conditions that mimic physiological or pathological states relevant to the research question.

How can researchers address the potential confounding effects of PKM1 upregulation in PKM2 knockout models?

The compensatory upregulation of PKM1 in PKM2 knockout models presents a significant confounding factor that must be addressed in experimental design and data interpretation:

  • Generate double knockout models:

    • Create conditional double knockout mice where both PKM1 and PKM2 can be deleted in specific tissues

    • Compare phenotypes between PKM2-only knockouts and PKM1/PKM2 double knockouts to isolate PKM2-specific effects

  • Use acute knockdown approaches:

    • Employ inducible shRNA or CRISPR-Cas9 systems to achieve acute PKM2 depletion before substantial PKM1 compensation occurs

    • Compare acute versus chronic PKM2 deletion effects to differentiate direct consequences from adaptive responses

  • Perform rescue experiments:

    • Re-express PKM2 in knockout cells to determine which phenotypes are reversible

    • Use PKM2 mutants with altered enzymatic activity or nuclear localization to dissect the mechanisms responsible for specific phenotypes

  • Employ PKM2-specific inhibitors:

    • Use small molecules that inhibit PKM2 activity or nuclear translocation without affecting PKM1

    • Compare pharmacological inhibition with genetic deletion to identify PKM2-specific functions

  • Conduct thorough metabolic characterization:

    • Perform comprehensive metabolomics to identify metabolic differences between wild-type, PKM2 knockout, and PKM1-expressing cells

    • Use stable isotope tracing to determine if PKM1 upregulation functionally compensates for PKM2 loss in terms of carbon flux

What are the potential non-glycolytic functions of PKM2 in mouse models and how can they be studied?

PKM2 has been proposed to have several non-glycolytic functions, though some remain controversial. To investigate these functions in mouse models, researchers should consider the following approaches:

  • Nuclear translocation and transcriptional regulation:

    • Use nuclear/cytoplasmic fractionation followed by Western blotting to assess PKM2 nuclear localization under various conditions

    • Perform chromatin immunoprecipitation (ChIP) assays to identify PKM2 binding to specific gene promoters

    • Use small molecules that specifically inhibit PKM2 nuclear translocation to distinguish between metabolic and nuclear functions

  • Protein kinase activity:

    • Assess phosphorylation of reported PKM2 substrates in wild-type versus PKM2 knockout tissues

    • Perform in vitro kinase assays using purified PKM2 and potential substrates

    • Generate knock-in mice expressing PKM2 mutants with impaired kinase activity but intact glycolytic function

  • Protein-protein interactions:

    • Conduct co-immunoprecipitation experiments to identify PKM2 binding partners in different tissues

    • Perform proximity ligation assays to visualize protein interactions in situ

    • Use mass spectrometry-based interactome analysis to comprehensively map PKM2 protein interactions

  • Redox regulation:

    • Measure cellular redox status in PKM2-deficient versus wild-type cells

    • Assess oxidative modifications of PKM2 in response to oxidative stress

    • Investigate how PKM2 deletion affects antioxidant responses in different tissues

  • Extracellular functions:

    • Examine if PKM2 is secreted from specific cell types under certain conditions

    • Investigate potential receptor-mediated signaling by extracellular PKM2

    • Assess how circulation levels of PKM2 correlate with disease progression in mouse models

By investigating these non-glycolytic functions, researchers can gain a more comprehensive understanding of PKM2's multifaceted roles beyond its canonical function in glycolysis, potentially explaining the diverse phenotypes observed in different PKM2 knockout models.

How can researchers resolve contradictory findings between different PKM2 knockout mouse models?

The disparate phenotypes observed across different PKM2 knockout mouse models—from protection against inflammatory diseases to promotion of liver cancer—require careful methodological approaches to resolve:

  • Standardize genetic backgrounds:

    • Back-cross mouse lines to identical genetic backgrounds

    • Use littermate controls to minimize strain-specific differences

    • Consider generating new knockouts on multiple genetic backgrounds to test for strain-dependent effects

  • Control for developmental timing:

    • Compare inducible knockout models (activated in adult mice) with germline knockouts to distinguish between developmental and adult functions of PKM2

    • Perform temporal studies to determine how knockout phenotypes evolve over time

  • Carefully define cell type specificity:

    • Use multiple validation techniques to confirm the specificity and completeness of conditional knockouts

    • Consider potential effects of PKM2 deletion in unintended cell types due to promiscuous Cre expression

    • Perform single-cell analyses to assess heterogeneity in knockout efficiency

  • Address compensatory mechanisms:

    • Thoroughly characterize PKM1 upregulation across different models

    • Perform transcriptomic and proteomic analyses to identify model-specific compensatory pathways

    • Use acute deletion systems to minimize time for compensatory adaptations

  • Consider context-dependent functions:

    • Design experiments that directly compare PKM2 functions under different metabolic conditions

    • Investigate how diet, inflammation, or other stressors modify phenotypes in PKM2 knockout models

    • Examine interaction effects between PKM2 and other genes/pathways that might explain context-dependent outcomes

  • Perform cross-laboratory validation:

    • Establish collaborations to test key findings across different laboratories

    • Share mouse models and standardized protocols to enhance reproducibility

    • Conduct meta-analyses of published data to identify consistent patterns across studies

By implementing these methodological approaches, researchers can reconcile seemingly contradictory findings and develop a more nuanced understanding of how PKM2 functions in different physiological and pathological contexts.

Product Science Overview

Introduction

Tumour Type M2 Pyruvate Kinase (PKM2) is a crucial enzyme in the glycolytic pathway, which is responsible for the conversion of phosphoenolpyruvate (PEP) to pyruvate, producing ATP in the process . PKM2 is one of the four isoforms of pyruvate kinase, the others being PKL, PKR, and PKM1 . This enzyme is particularly significant in cancer research due to its unique role in tumor metabolism and its potential as a diagnostic marker.

Isoforms of Pyruvate Kinase

Pyruvate kinase exists in four isoforms, each with tissue-specific expression:

  • PKL: Found in the liver.
  • PKR: Present in red blood cells.
  • PKM1: Expressed in tissues with high energy demands such as the brain, skeletal muscle, and heart.
  • PKM2: Found in early embryonic cells and proliferating cells, including tumor cells .
Role of PKM2 in Tumor Cells

PKM2 is predominantly expressed in tumor cells and exists mainly in its dimeric form, which has a lower affinity for PEP compared to its tetrameric form . This dimeric form, often referred to as Tumor M2-PK, is associated with the metabolic reprogramming of cancer cells, known as the Warburg effect . This effect describes how cancer cells preferentially produce energy through aerobic glycolysis rather than oxidative phosphorylation, even in the presence of sufficient oxygen .

Metabolic and Non-Metabolic Functions

In addition to its metabolic role, PKM2 has non-metabolic functions that contribute to tumorigenesis. It can translocate to the nucleus and act as a protein kinase, phosphorylating various targets and influencing multiple physiological processes . This dual functionality makes PKM2 a critical player in cancer cell metabolism and proliferation.

Diagnostic and Therapeutic Potential

The unique properties of PKM2, particularly its dimeric form in tumor cells, make it a valuable biomarker for cancer diagnosis and monitoring . Quantifying Tumor M2-PK in plasma and stool samples can aid in the early detection of tumors and in tracking the effectiveness of cancer therapies . Furthermore, targeting PKM2’s activity and its regulatory mechanisms holds promise for developing novel cancer treatments .

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 2025 TheBiotek. All Rights Reserved.